US20230124705A1 - Solid forms, pharmaceutical compositions and preparation of heteroaromatic macrocyclic ether compounds - Google Patents

Solid forms, pharmaceutical compositions and preparation of heteroaromatic macrocyclic ether compounds Download PDF

Info

Publication number
US20230124705A1
US20230124705A1 US17/957,725 US202217957725A US2023124705A1 US 20230124705 A1 US20230124705 A1 US 20230124705A1 US 202217957725 A US202217957725 A US 202217957725A US 2023124705 A1 US2023124705 A1 US 2023124705A1
Authority
US
United States
Prior art keywords
compound
solid form
solvent
formula
xrpd pattern
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/957,725
Other languages
English (en)
Inventor
Sibao Chen
Christopher G. F. COOPER
Baudouin Gerard
Joshua Courtney Horan
Jason T. KROPP
Benjamin Stephen LANE
David James Pearson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nuvalent Inc
Original Assignee
Nuvalent Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nuvalent Inc filed Critical Nuvalent Inc
Priority to US17/957,725 priority Critical patent/US20230124705A1/en
Assigned to CAMBREX CORPORATION reassignment CAMBREX CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PEARSON, DAVID JAMES
Assigned to NUVALENT, INC. reassignment NUVALENT, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HORAN, JOSHUA COURTNEY, LANE, BENJAMIN STEPHEN, GERARD, BAUDOUIN, KROPP, JASON T.
Assigned to NUVALENT, INC. reassignment NUVALENT, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CAMBREX CORPORATION
Assigned to NUVALENT, INC. reassignment NUVALENT, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: COOPER, Christopher G. F., CHEN, Sibao
Publication of US20230124705A1 publication Critical patent/US20230124705A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/22Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2059Starch, including chemically or physically modified derivatives; Amylose; Amylopectin; Dextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • RTKs Receptor tyrosine kinases
  • Many RTKs are proto-oncogenes; aberrant RTK activity can drive cell survival, growth and proliferation leading to cancer and related disorders. This aberrant kinase activity can be caused by mutations such as activating mutations in the kinase domain, gene rearrangements that result in fusion proteins containing the intact kinase domain, amplification and other means.
  • RTK proto-oncogenes include ROS1, anaplastic lymphoma kinase (ALK), NTRK1 (encodes TRKA), NTRK2 (encodes TRKB), and NTRK3 (encodes TRKC).
  • ROS1 is an RTK proto-oncogene, with ROS1 rearrangements detected in non-small cell lung cancer (NSCLC), glioblastoma, inflammatory myofibroblastic tumor (IMT), cholangiocarcinoma, ovarian cancer, gastric cancer, colorectal cancer, angiosarcoma, and spitzoid melanoma.
  • NSCLC non-small cell lung cancer
  • IMT inflammatory myofibroblastic tumor
  • cholangiocarcinoma cholangiocarcinoma
  • ovarian cancer gastric cancer
  • colorectal cancer colorectal cancer
  • angiosarcoma angiosarcoma
  • spitzoid melanoma spitzoid melanoma.
  • ROS1 fusion partner genes observed in NSCLC include SLC34A2, CD74, TPM3, SDC4, EZR, LRIG3, KDELR2, CEP72, CLTL, CTNND2, GOPC, GPRC6A, LIAMA1, LRIG3, MSN, MYO5C, OPRM1, SLC6A17 (putative), SLMAP, SRSF6, TFG, TMEM106B, TPD52L1, ZCCHC8 and CCDC6.
  • fusion partners include CAPRIN1, CEP85L, CHCHD3, CLIP1 (putative), EEF1G, KIF21A (putative), KLC1, SART3, ST13 (putative), TRIM24 (putative), ERC1, FIPIL1, HLAA, KIAA1598, MYO5A, PPFIBP1, PWWP2A, FN1, YWHAE, CCDC30, NCOR2, NFKB2, APOB, PLG, RBP4, and GOLGB1.
  • ALK is an RTK proto-oncogene, with ALK rearrangements detected in many cancers, including NSCLC, anaplastic large cell lymphoma (ALCL), IMT, diffuse large B-cell lymphoma (DLBCL), esophageal squamous cell carcinoma (ESCC), renal medullary carcinoma, renal cell carcinoma, breast cancer, colon cancer, serous ovarian carcinoma, papillary thyroid cancer, and spitzoid tumors, and ALK activating mutations detected in neuroblastoma.
  • Oncogenic ALK gene fusions contain the kinase domain of ALK (3′ region) fused to the 5′ region of more than 20 different partner genes, the most common being EML4 in NSCLC and NPM in ALCL.
  • NTRK1, NTRK2 and NTRK3 are RTK proto-oncogenes that encode TRK-family kinases, with NTRK1, NTRK2 and NTRK3 chromosomal rearrangements detected at low frequency in many cancers.
  • TRK inhibition particularly in the central nervous system (CNS) has been associated with adverse reactions, including dizziness/ataxia/gait disturbance, paraesthesia, weight gain and cognitive changes.
  • ROS1 and ALK have substantial deficiencies. These deficiencies may represent one or more of the following: associated TRK inhibition, limited CNS activity, and inadequate activity against resistance mutations. Treatment of ROS1-positive or ALK-positive patients accompanied by TRK inhibition is associated with adverse reactions, particularly in the CNS, including dizziness/ataxia/gait disturbance, paraesthesia, weight gain and cognitive changes.
  • CNS-penetrant and TRK-sparing inhibitors of the wild type ROS1 kinase domain and ROS1 with acquired resistance mutations occurring either individually or in combination, including G2032R, D2033N, S1986F, S1986Y, L2026M, L1951R, E1935G, L1947R, G1971E, E1974K, L1982F, F2004C, F2004V, E2020K, C2060G, F2075V, V2089M, V2098I, G2101A, D2113N, D2113G, L2155S, L2032K, and L2086F.
  • ALK drug resistance mutations including G1202R, L1196M, G1269A, C1156Y, I1171T, I1171N, I1171S, F1174L, V1180L, S1206Y, E1210K, 1151Tins, F1174C, G1202del, D1203N, S1206Y, S1206C, L1152R, L1196Q, L1198P, L1198F, R1275Q, L1152P, C1156T, and F1245V.
  • solid forms comprising a compound of formula (I) (also referred as Compound 1) or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof:
  • the solid form is a crystalline form. In other embodiments, the solid form is an amorphous form. In some embodiments, the solid form is a solid form of a compound of formula (I). In some embodiments, the solid form is a solid form of a free base of a compound of formula (I). In some embodiments, the solid form is a solid form of a salt of a compound of formula (I). In some embodiments, the solid form is a crystalline form of a free base of a compound of formula (I). In some embodiments, the solid form is a crystalline form of a salt of a compound of formula (I).
  • Also provided herein are methods of treating cancer comprising administering a therapeutically effective amount of a solid form of a compound of formula (I) provided herein to a subject in need thereof.
  • compositions comprising a solid form of a compound of formula (I) and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition comprises a solid form of a free base of a compound of formula (I).
  • the pharmaceutical composition comprises a solid form of a salt of a compound of formula (I).
  • step 1.0 cyclizing a compound of Formula (II):
  • compositions comprising a compound of formula (I):
  • Also provided herein are methods of treating cancer comprising administering a therapeutically effective amount of the pharmaceutical composition provided herein.
  • FIG. 1 is a representative X-ray powder diffraction (XRPD) pattern of Form 1 of free base of Compound 1.
  • FIG. 2 is a representative integrated thermal gravimetric analysis (TGA) and differential thermal analysis (DTA) thermograms for Form 1 of free base of Compound 1.
  • FIG. 3 is a representative and differential scanning calorimetry (DSC) thermogram of Form 1 of free base of Compound 1.
  • FIG. 4 is a representative dynamic vapor sorption (DVS) isotherm of Form 1 of free base of Compound 1.
  • FIG. 5 is a representative fourier-transform infrared spectrum (FT-IR) spectrum of Form 1 of free base of Compound 1.
  • FIG. 6 is a representative depiction of the unit cell b axis of single-crystal X-ray diffraction studies of Form 1 of free base of Compound 1.
  • FIG. 7 is a representative XRPD pattern of Form 2 of free base of Compound 1.
  • FIG. 8 is a representative overlay of TGA and differential thermal analysis (DTA) thermograms for Form 2 of free base of Compound 1.
  • FIG. 9 is another representative overlay of TGA and differential thermal analysis (DTA) thermograms for of Form 2 of free base of Compound 1.
  • FIG. 10 is a representative DSC thermogram of Form 2 of free base of Compound 1.
  • FIG. 11 is a representative FT-IR spectrum of Form 2 of free base of Compound 1.
  • FIG. 12 is a representative XRPD pattern of Form 3 of free base of Compound 1.
  • FIG. 13 is a representative overlay of TGA and DTA thermograms for Form 3 of free base of Compound 1.
  • FIG. 14 is a representative XRPD pattern of Form 4 of free base of Compound 1.
  • FIG. 15 is a representative overlay of TGA and DTA thermograms for Form 4 of free base of Compound 1.
  • FIG. 16 is a representative XRPD pattern of Form 5 of free base of Compound 1.
  • FIG. 17 is a representative XRPD pattern of Form 6 of free base of Compound 1.
  • FIG. 18 is a representative overlay of TGA and DTA thermograms for Form 6 of free base of Compound 1.
  • FIG. 19 is a representative XRPD pattern of Form 7 of free base of Compound 1.
  • FIG. 20 is a representative DSC thermogram of Form 7 of free base of Compound 1.
  • FIG. 21 is a representative DVS isotherm of Form 7 of free base of Compound 1.
  • FIG. 22 is a representative FT-IR spectrum of Form 7 of free base of Compound 1.
  • FIG. 23 is a representative depiction of the unit cell from single-crystal X-ray diffraction studies of Form 7 of free base of Compound 1.
  • FIG. 24 is a representative XRPD pattern of Form A of a besylate salt of Compound 1.
  • FIG. 25 is a representative overlay of TGA and DSC thermograms for Form A of a besylate salt of Compound 1.
  • FIG. 26 is a representative DSC thermogram for Form A of a besylate salt of Compound 1.
  • FIG. 27 is a representative DVS isotherm of Form A of a besylate salt of Compound 1.
  • FIG. 28 is a representative XRPD pattern of Form A of a phosphate salt of Compound 1.
  • FIG. 29 is a representative overlay of TGA and DSC thermograms of Form A of a phosphate salt of Compound 1.
  • chemical structures are disclosed with a corresponding chemical name. In case of conflict, the chemical structure controls the meaning, rather than the name.
  • the terms “comprising” and “including” can be used interchangeably.
  • the terms “comprising” and “including” are to be interpreted as specifying the presence of the stated features or components as referred to, but does not preclude the presence or addition of one or more features, or components, or groups thereof. Additionally, the terms “comprising” and “including” are intended to include examples encompassed by the term “consisting of”. Consequently, the term “consisting of” can be used in place of the terms “comprising” and “including” to provide for more specific embodiments of the invention.
  • stereoisomers refer to the various stereoisomeric forms of a compound that comprises one or more asymmetric centers or stereohindrance in the structure.
  • a stereoisomer is an enantiomer, a mixture of enantiomers, an atropisomer, or a tautomer thereof.
  • the compounds described herein can be in the form of an individual enantiomer, diastereomer or geometric isomer (e.g. an atropisomer), or can be in the form of a mixture of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomer.
  • compounds provided herein may be atropisomers.
  • Atropisomers are stereoisomers arising because of hindered rotation about a single bond, where energy differences due to steric strain or other contributors create a barrier to rotation that is high enough to allow for isolation of individual conformers.
  • Stereoisomers can be isolated from mixtures by methods known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts; or preferred isomers can be prepared by asymmetric syntheses. See, for example, Jacques et al., Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Wilen et al., Tetrahedron 33:2725 (1977); Eliel, E. L.
  • compounds provided herein may be racemic. In certain embodiments, compounds provided herein may be enriched in one enantiomer. For example, a compound provided herein may have greater than about 30% ee, about 40% ee, about 50% ee, about 60% ee, about 70% ee, about 80% ee, about 90% ee, or even about 95% or greater ee. In certain embodiments, compounds provided herein may have more than one stereocenter. In certain such embodiments, compounds provided herein may be enriched in one or more diastereomer. For example, a compound provided herein may have greater than about 30% de, about 40% de, about 50% de, about 60% de, about 70% de, about 80% de, about 90% de, or even about 95% or greater de.
  • the therapeutic preparation may be enriched to provide predominantly one enantiomer of a compound.
  • An enantiomerically enriched mixture may comprise, for example, at least about 60 mol percent of one enantiomer, or more particularly at least about 75, about 90, about 95, or even about 99 mol percent.
  • the compound enriched in one enantiomer is substantially free of the other enantiomer, wherein substantially free means that the substance in question makes up less than about 10%, or less than about 5%, or less than about 4%, or less than about 3%, or less than about 2%, or less than about 1% as compared to the amount of the other enantiomer, e.g., in the composition or compound mixture.
  • composition or compound mixture contains about 98 grams of a first enantiomer and about 2 grams of a second enantiomer, it would be said to contain about 98 mol percent of the first enantiomer and only about 2% of the second enantiomer.
  • the therapeutic preparation may be enriched to provide predominantly one diastereomer of a compound.
  • a diastereomerically enriched mixture may comprise, for example, at least about 60 mol percent of one diastereomer, or more particularly at least about 75, about 90, about 95, or even about 99 mol percent.
  • a moiety in a compound exists as a mixture of tautomers.
  • a “tautomer” is a structural isomer of a moiety or a compound that readily interconverts with another structural isomer.
  • a pyrazole ring has two tautomers:
  • subject to which administration is contemplated includes, but is not limited to, humans (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult or senior adult)) and/or other primates (e.g., cynomolgus monkeys, rhesus monkeys); mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, goats, cats, and/or dogs; and/or birds, including commercially relevant birds such as chickens, ducks, geese, quail, and/or turkeys.
  • the subject is a human.
  • the subject is a human adult at least of 40 years old. In certain embodiments, the subject is a human adult at least of 50 years old. In certain embodiments, the subject is a human adult at least of 60 years old. In certain embodiments, the subject is a human adult at least of 70 years old. In certain embodiments, the subject is a human adult at least of 18 years old or at least of 12 years old.
  • a human subject to which administration of a therapeutic e.g., a compound as described herein
  • a patient a human subject to which administration of a therapeutic (e.g., a compound as described herein) is contemplated in order to treat, prevent or manage a disease, disorder, or condition, or symptoms thereof, is also called a “patient”.
  • a therapeutic that “prevents” a disorder or condition refers to a compound that, in a statistical sample, reduces the occurrence of the disorder or condition in the treated sample relative to an untreated control sample, or delays the onset or reduces the severity of one or more symptoms of the disorder or condition relative to the untreated control sample.
  • effects are also called “prophylactic” effects.
  • prevention and preventing refer to an approach for obtaining beneficial or desired results including, but not limited, to prophylactic benefit.
  • a therapeutic can be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • a therapeutic is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the subject) for prophylactic benefit (e.g., it protects the subject against developing the unwanted condition).
  • treatment refers to therapeutic or palliative measures.
  • Beneficial or desired clinical results include, but are not limited to, alleviation, in whole or in part, of symptoms associated with a disease or disorder or condition, diminishment of the extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state (e.g., one or more symptoms of the disease), and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • “treatment” comprises administration of a therapeutic after manifestation of the unwanted condition (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • the term “managing” encompasses preventing the recurrence of the particular disease or disorder in a patient who had suffered from it, lengthening the time a patient who had suffered from the disease or disorder remains in remission, reducing mortality rates of the patients, and/or maintaining a reduction in severity or avoidance of a symptom associated with the disease or condition being managed.
  • an “effective amount”, as used herein, refers to an amount that is sufficient to achieve a desired biological effect.
  • a “therapeutically effective amount”, as used herein, refers to an amount that is sufficient to achieve a desired therapeutic effect.
  • a therapeutically effective amount can refer to an amount that is sufficient to improve at least one sign or symptom of cancer.
  • a “response” to a method of treatment can include a decrease in or amelioration of negative symptoms, a decrease in the progression of a disease or symptoms thereof, an increase in beneficial symptoms or clinical outcomes, a lessening of side effects, stabilization of disease, partial or complete remedy of disease, among others.
  • the term “relapsed” refers to a disorder, disease, or condition that responded to prior treatment (e.g., achieved a complete response) then had progression.
  • the prior treatment can include one or more lines of therapy.
  • the term “refractory” refers to a disorder, disease, or condition that has not responded to prior treatment that can include one or more lines of therapy.
  • Crystal refers to a homogeneous solid formed by a repeating, three-dimensional pattern of atoms, ions or molecules having fixed distances between constituent parts. The unit cell is the simplest repeating unit in this pattern. Notwithstanding the homogenous nature of an ideal crystal, a perfect crystal rarely, if ever, exists. “Crystalline,” as used herein, encompasses crystalline forms that include crystalline defects, for example, crystalline defects commonly formed by manipulating (e.g., preparing, purifying) the crystalline forms described herein. A person skilled in the art is capable of determining whether a sample of a compound is crystalline notwithstanding the presence of such defects.
  • Crystalline forms can be characterized by analytical methods such as x-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), nuclear magnetic resonance spectroscopy (NMR), single crystal x-ray diffraction, Raman spectroscopy, Fourier transform infrared spectroscopy (FTIR) and/or any other suitable analytical techniques.
  • analytical methods such as x-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), nuclear magnetic resonance spectroscopy (NMR), single crystal x-ray diffraction, Raman spectroscopy, Fourier transform infrared spectroscopy (FTIR) and/or any other suitable analytical techniques.
  • solvate refers to a crystalline form of a molecule, atom, and/or ions that further comprises molecules of a solvent or solvents incorporated into the crystalline lattice structure.
  • the solvent molecules in the solvate may be present in a regular arrangement and/or a non-ordered arrangement.
  • the solvate may comprise either a stoichiometric or nonstoichiometric amount of the solvent molecules.
  • a solvate with a nonstoichiometric amount of solvent molecules may result from partial loss of solvent from the solvate.
  • Solvates may occur as dimers or oligomers comprising more than one molecule or Compound ABC within the crystalline lattice structure.
  • amorphous refers to a solid form of a molecule, atom, and/or ions that is not crystalline.
  • the term “amorphous form” describes a disordered solid form, i.e., a solid form lacking long range crystalline order. An amorphous solid does not display a definitive X-ray diffraction pattern.
  • an amorphous form of a substance may be substantially pure of other amorphous forms and/or crystal forms.
  • solid form and related terms refer to a physical form which is not predominantly in a liquid or a gaseous state. Solid forms may be crystalline, amorphous or mixtures thereof. As used herein and unless otherwise specified, the term “crystal forms” and related terms refer to solid forms that are crystalline. Crystal forms include, but are not limited to, non-solvates, non-hydrates, solvates, hydrates, and other molecular complexes, as well as salts, solvates of salts, hydrates of salts, and other molecular complexes of salts thereof.
  • a solid form or crystal form of a substance may be substantially free of amorphous forms and/or other solid forms and/or crystal forms.
  • a solid form and/or crystal form of a substance may contain less than about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50% of one or more amorphous forms and/or other solid forms and/or crystal forms on a weight basis.
  • a solid form or crystal form of a substance may be physically and/or chemically pure.
  • a solid form or crystal form of a substance may be about 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91% or 90% physically and/or chemically pure. In certain embodiments, a solid form or crystal form of a substance may be at least about 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91% or 90% physically and/or chemically pure. In one embodiment, a solid form or crystal form of a substance is about 97.0% physically and/or chemically pure on a weight basis. In one embodiment, a solid form or crystal form of a substance is about 98.0% physically and/or chemically pure on a weight basis.
  • a solid form or crystal form of a substance is about 99.0% physically and/or chemically pure on a weight basis. In one embodiment, a solid form or crystal form of a substance is about 99.9% physically and/or chemically pure on a weight basis. In certain embodiments, a solid form or crystal form may be substantially chemically pure and/or substantially physically pure.
  • “Substantially pure,” when used without further qualification, means the compound has a purity greater than 90 weight percent, for example, greater than 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 weight percent, and also including a purity equal to about 100 weight percent, based on the weight of the compound.
  • the remaining material may comprise other form(s) of the compound and/or reaction impurities and/or processing impurities arising from its preparation. Purity can be assessed using techniques known in the art, for example, using an HPLC assay.
  • “Substantially pure” can also be qualified. If the compound is “substantially pure” with respect to the presence of chemical impurities (e.g. reaction impurities and/or processing impurities arising from its preparation), it can be referred to as “substantially chemically pure”. If the compound is “substantially pure” with respect to the presence of the other enantiomer, it can be referred to as “substantially enantiomerically pure”. In some embodiments, the compound (e.g. Compound 1) is substantially enantiomerically pure with the other enantiomer (e.g.
  • the S enanontiomer present less than 10%, less than 5%, less than 3%, less than 1%, less than 0.5%, or less than 0.1% by weight. If the compound is “substantially pure” with respect to the presence of other physical forms of the compound having the indicated structure, it can be referred to as “substantially physically pure”. When qualified, “substantially pure” means that the indicated compound contains less than 10%, less than 5%, less than 3%, less than 1%, less than 0.5%, or less than 0.1% by weight of the indicated impurity. In certain embodiments, the solid form of Compound 1 is substantially pure (e.g.
  • the solid form of Compound 1 has the purity of at least about 95 wt %. In certain embodiments, the solid form of Compound 1 is substantially enantiomerically pure (e.g. having the enantiomeric purity of at least about 98.0 wt %, at least about 99.0 wt %, at least about 99.5 wt %, or at least about 99.9 wt %).
  • the solid form of Compound 1 has the enantiomeric purity of at least about 99.5 wt %.
  • the pharmaceutical composition comprising Compound 1 has the purity of at least about 95 wt %.
  • the pharmaceutical composition comprising Compound 1 has the purity of at least about 96 wt %.
  • the pharmaceutical composition comprising Compound 1 has the purity of at least about 97 wt %.
  • the pharmaceutical composition comprising Compound 1 has the purity of at least about 98 wt %.
  • the pharmaceutical composition comprising Compound 1 has the purity of at least about 99 wt %.
  • the pharmaceutical composition comprising Compound 1 has the purity of at least about 95 wt % over 12 months.
  • Solid forms may exhibit distinct physical characterization data that are unique to a particular solid form, such as the crystal forms described herein. These characterization data may be obtained by various techniques known to those skilled in the art. The data provided by these techniques may be used to identify a particular solid form. For example, an XRPD pattern, DSC thermogram or TGA thermal curve that “matches” or, interchangeably, is “substantially in accordance” with one or more figures herein showing an XRPD pattern or DSC thermogram or TGA thermal curve, respectively, is one that would be considered by one skilled in the art to represent the same single crystalline form of the compound as the sample of the compound that provided the pattern or thermogram or thermal curve of one or more figures provided herein.
  • an XRPD pattern or DSC thermogram or TGA thermal curve that matches or is substantially in accordance may be identical to that of one of the figures or, more likely, may be somewhat different from one or more of the figures.
  • an XRPD pattern that is somewhat different from one or more of the figures may not necessarily show each of the lines of the diffraction pattern presented herein and/or may show a slight change in appearance or intensity of the lines or a shift in the position of the lines. These differences typically result from differences in the conditions involved in obtaining the data or differences in the purity of the sample used to obtain the data.
  • a person skilled in the art is capable of determining if a sample of a crystalline compound is of the same form as or a different form from a form disclosed herein by comparison of the XRPD pattern or DSC thermogram or TGA thermal curve of the sample and the corresponding XRPD pattern or DSC thermogram or TGA thermal curve disclosed herein.
  • the terms “about” and “approximately,” when used in connection with a numeric value or a range of values which is provided to characterize a particular solid form e.g., a specific temperature or temperature range, such as, for example, that describing a melting, dehydration, desolvation or glass transition temperature; a mass change, such as, for example, a mass change as a function of temperature or humidity; a solvent or water content, in terms of, for example, mass or a percentage; or a peak position, such as, for example, in analysis by IR or Raman spectroscopy or XRPD; indicate that the value or range of values may deviate to an extent deemed reasonable to one of ordinary skill in the art while still describing the particular solid form.
  • the terms “about” and “approximately,” when used in this context, indicate that the numeric value or range of values may vary within 25%, 20%, 15%, 10%, 9% 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1.5%, 1%, 0.5%, or 0.25% of the recited value or range of values.
  • the value of XRPD peak position may vary by up to ⁇ 0.2 degrees 2 ⁇ while still describing the particular XRPD peak.
  • he value of XRPD peak position may vary by up to +0.1 degrees 2 ⁇ .
  • he value of XRPD peak position may vary by up to +0.05 degrees 2 ⁇ .
  • pharmaceutically acceptable salt refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of subjects without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences (1977) 66:1-19.
  • pharmaceutically acceptable salts include, but are not limited to, alkyl, dialkyl, trialkyl or tetra-alkyl ammonium salts.
  • pharmaceutically acceptable salts include, but are not limited to, L-arginine, benenthamine, benzathine, betaine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)ethanol, ethanolamine, ethylenediamine, N-methylglucamine, hydrabamine, 1H-imidazole, lithium, L-lysine, magnesium, 4-(2-hydroxyethyl)morpholine, piperazine, potassium, 1-(2-hydroxyethyl)pyrrolidine, sodium, triethanolamine, tromethamine, and zinc salts.
  • pharmaceutically acceptable salts include, but are not limited to, Na, Ca, K, Mg, Zn or other metal salts.
  • the pharmaceutically acceptable acid addition salts can also exist as various solvates, such as with water, methanol, ethanol, dimethylformamide, and the like. Mixtures of such solvates can also be prepared.
  • the source of such solvate can be from the solvent of crystallization, inherent in the solvent of preparation or crystallization, or adventitious to such solvent.
  • anionic salts include, but are not limited to, acetate, aspartate, benzenesulfonate, benzoate, besylate, bicarbonate, bitartrate, bromide, camsylate, carbonate, chloride, citrate, decanoate, edetate, esylate, fumarate, gluceptate, gluconate, glutamate, glycolate, hexanoate, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandelate, mesylate, methylsulfate, mucate, napsylate, nitrate, octanoate, oleate, pamoate, pantothenate, phosphate, polygalacturonate, propionate, salicylate, stearate, acetate, succinate, sulfate, tartrate, teoclate, and tosylate.
  • the term “enantiomerically pure” refers to a composition comprising an enantiomeric excess of at least about 50%, at least about 75%, at least about 90%, at least about 95%, or at least about 99% of one enantiomer of a compound having one or more chiral center(s).
  • the composition may be “substantially enantiomerically pure”, which refers to preparations of compositions which have at least about 85% by weight of one enantiomer relative to the other enantiomer of a compound, such as at least about 90% by weight, and further such as at least 95% by weight.
  • compositions provided herein comprise an enantiomeric excess of at least about 90% by weight of one enantiomer of the compound. In other embodiments, the compositions comprises an enantiomeric excess of at least about 95%, at least about 98%, or at least about 99% by weight of one enantiomer of the compound.
  • process(es) refers to the methods provided herein which are useful for preparing a compound as described herein or a solid form thereof (e.g. a crystalline form, partically crystalline form, or an oxyphous form) provided herein.
  • a solid form thereof e.g. a crystalline form, partically crystalline form, or an oxyphous form
  • Modifications to the methods provided herein e.g., starting materials, reagents, protecting groups, solvents, temperatures, reaction times, purification
  • the technical teaching of one embodiment provided herein can be combined with that disclosed in any other embodiments provided herein.
  • the term “adding,” “reacting,” “treating,” or the like means contacting one reactant, reagent, solvent, catalyst, reactive group or the like with another reactant, reagent, solvent, catalyst, reactive group or the like.
  • Reactants, reagents, solvents, catalysts, reactive group or the like can be added individually, simultaneously or separately and can be added in any order.
  • Reactants, reagents, solvents, catalysts, reactive group or the like can each respectively be added in one portion, which may be delivered all at once or over a period of time, or in discrete portions, which also may be delivered all at once or over a period of time. They can be added in the presence or absence of heat and can optionally be added under an inert atmosphere.
  • “Reacting” can refer to in situ formation or intramolecular reaction where the reactive groups are in the same molecule.
  • the term “combining” refers to bringing one or more chemical entities into association with another one or more chemical entities.
  • Combining includes the processes of adding one or more compounds to a solid, liquid or gaseous mixture of one or more compounds (the same or other chemical entities), or a liquid solution or multiphasic liquid mixture.
  • the act of combining includes the process or processes of one or more compounds reacting (e.g., bond formation or cleavage; salt formation, solvate formation, chelation, or other non-bond altering association) with one or more compounds (the same or other chemical entities).
  • the act of combining can include alteration of one or more compounds, such as by isomerization (e.g., tautomerization, resolution of one isomer from another, or racemization).
  • the term “transforming” refers to subjecting the compound at hand to reaction conditions suitable to effect the formation of the desired compound at hand.
  • the term “recovering” includes, but is not limited to, the action of obtaining one or more compounds by collection during and/or after a process step as disclosed herein, and the action of obtaining one or more compounds by separation of one or more compounds from one or more other chemical entities during and/or after a process step as disclosed herein.
  • the term “collection” refers to any action(s) known in the art for this purpose, including, but not limited to, filtration, decanting a mother liquor from a solid to obtain one or more compounds, and evaporation of liquid media in a solution or other mixture to afford a solid, oil, or other residue that includes one or more compounds.
  • the solid can be crystalline, acrystalline, partially crystalline, or amorphous, a powder, granular, of varying particle sizes, of uniform particle size, among other characteristics known in the art.
  • An oil can vary in color and viscosity, and include one or more solid forms as a heterogeneous mixture, among other characteristics known in the art.
  • separation refers to any action(s) known in the art for this purpose, including, but not limited to, isolating one or more compounds from a solution or mixture using, for example, seeded or seedless crystallization or other precipitation techniques (e.g., adding an anti-solvent to a solution to induce compound precipitation; heating a solution, then cooling to induce compound precipitation; scratching the surface of a solution with an implement to induce compound precipitation), and distillation techniques. Recovering one or more compounds can involve preparation of a salt, solvate, hydrate, chelate or other complexes of the same, then collecting or separating as described above.
  • seeded or seedless crystallization or other precipitation techniques e.g., adding an anti-solvent to a solution to induce compound precipitation; heating a solution, then cooling to induce compound precipitation; scratching the surface of a solution with an implement to induce compound precipitation
  • Recovering one or more compounds can involve preparation of a salt, solvate, hydrate, chelate or other complex
  • catalyst precursor refers to a chemical composition wherein one or more components of an active catalyst (e.g. metal center and supporting ligand) are added to the reaction mixture such that formation of an active catalyst occurs in situ.
  • an active catalyst e.g. metal center and supporting ligand
  • a cataCXium A ligated palladium catalyst may be formed in situ by adding a catalyst precursor comprising a palladium source (e.g. Pd(OAc) 2 ) and a source of cataCXium A (e.g. cataCXium A).
  • a palladium source e.g. Pd(OAc) 2
  • a source of cataCXium A e.g. cataCXium A
  • catalyst includes, but is not limited to a chemical composition wherein more than one component of an active catalyst (e.g. metal center and supporting ligand) is added to a reaction mixture in the form of a single chemical entity (e.g. Pd(dppf)Cl 2 ), even if further activation and/or reaction in situ is required to produce an active catalyst.
  • active catalyst e.g. metal center and supporting ligand
  • solvent As used herein, and unless otherwise specified, the terms “solvent,” “organic solvent,” or “inert solvent” each mean a solvent inert under the conditions of the reaction being described. Unless specified to the contrary, for each gram of a limiting reagent, one cc (or mL) of solvent constitutes a volume equivalent (or “vol.”).
  • Potential pharmaceutical solids include crystalline solids and amorphous solids.
  • Amorphous solids are characterized by a lack of long-range structural order, whereas crystalline solids are characterized by structural periodicity.
  • the desired class of pharmaceutical solid depends upon the specific application; amorphous solids are sometimes selected on the basis of, e.g., an enhanced dissolution profile, while crystalline solids may be desirable for properties such as, e.g., physical or chemical stability (see, e.g., S. R. Vippagunta et al., Adv. Drug. Deliv. Rev ., (2001) 48:3-26; L. Yu, Adv. Drug. Deliv. Rev ., (2001) 48:27-42).
  • a change in solid form may affect a variety of physical and chemical properties, which may provide benefits or drawbacks in processing, formulation, stability and bioavailability, among other important pharmaceutical characteristics.
  • crystalline or amorphous, potential solid forms of a pharmaceutical compound may include single-component and multiple-component solids.
  • Single-component solids consist essentially of the pharmaceutical compound in the absence of other compounds. Variety among single-component crystalline materials may potentially arise from the phenomenon of polymorphism, wherein multiple three-dimensional arrangements exist for a particular pharmaceutical compound (see, e.g., S. R. Byrn et al., Solid State Chemistry of Drugs , (1999) SSCI, West Lafayette).
  • Crystalline solids comprising two or more ionic species are termed salts (see, e.g., Handbook of Pharmaceutical Salts: Properties, Selection and Use , P. H. Stahl and C. G. Wermuth, Eds., (2002), Wiley, Weinheim).
  • Additional types of multiple-component solids that may potentially offer other property improvements for a pharmaceutical compound or salt thereof include, e.g., hydrates, solvates, co-crystals and clathrates, among others (see, e.g., S. R. Byrn et al., Solid State Chemistry of Drugs , (1999) SSCI, West Lafayette).
  • Multiple-component crystal forms may potentially be susceptible to polymorphism, wherein a given multiple-component composition may exist in more than one three-dimensional crystalline arrangement.
  • the discovery of solid forms is of great importance in the development of a safe, effective, stable and marketable pharmaceutical compound.
  • solid forms provided herein are useful as active pharmaceutical ingredients for the preparation of formulations for use in animals or humans. Thus, embodiments herein encompass the use of these solid forms as a final drug product. Certain embodiments provide solid forms useful in making final dosage forms with improved properties, e.g., powder flow properties, compaction properties, tableting properties, stability properties, and excipient compatibility properties, among others, that are needed for manufacturing, processing, formulation and/or storage of final drug products. Certain embodiments herein provide pharmaceutical compositions comprising a single-component crystal form, and/or a multiple-component crystal form comprising the compound of formula (I) and a pharmaceutically acceptable excipient.
  • Solid form and related terms refer to a physical form which is not predominantly in a liquid or a gaseous state.
  • Solid forms may be crystalline or mixtures of crystalline and amorphous forms.
  • a “single-component” solid form comprising a particular compound consists essentially of that compound.
  • a “multiple-component” solid form comprising a particular compound comprises that compound and a significant quantity of one or more additional species, such as ions and/or molecules, within the solid form.
  • the solid forms provided herein may be crystalline or an intermediate form (e.g., a mixture of crystalline and amorphous forms). The crystal forms described herein, therefore, may have varying degrees of crystallinity or lattice order.
  • the solid forms described herein are not limited to any particular degree of crystallinity or lattice order, and may be 0-100% crystalline. Methods of determining the degree of crystallinity are known to those of ordinary skill in the art, such as those described in Suryanarayanan, R., X - Ray Powder Diffractometry , Physical Characterization of Pharmaceutical Solids, H. G. Brittain, Editor, Marcel Dekker, Murray Hill, N.J., 1995, pp. 187-199, which is incorporated herein by reference in its entirety. In some embodiments, the solid forms described herein are about 0, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100% crystalline.
  • Solid forms may exhibit distinct physical characterization data that are unique to a particular solid form, such as the crystal forms described herein.
  • These characterization data may be obtained by various techniques known to those skilled in the art, including for example X-ray powder diffraction, differential scanning calorimetry, thermal gravimetric analysis, and nuclear magnetic resonance spectroscopy. The data provided by these techniques may be used to identify a particular solid form.
  • One skilled in the art can determine whether a solid form is one of the forms described herein by performing one of these characterization techniques and determining whether the resulting data is “substantially similar” to the reference data provided herein, which is identified as being characteristic of a particular solid form.
  • Characterization data that is “substantially similar” to those of a reference solid form is understood by those skilled in the art to correspond to the same solid form as the reference solid form. In analyzing whether data is “substantially similar,” a person of ordinary skill in the art understands that particular characterization data points may vary to a reasonable extent while still describing a given solid form, due to, for example, experimental error and routine sample-to-sample analysis.
  • solid forms comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof:
  • the solid form comprising a compound of formula (I) can be a crystalline form, a partially crystalline form, or a mixture of crystalline form(s) and amorphous form(s).
  • a solid form comprising a crystalline form of a compound of formula (I).
  • the solid form comprises a salt, solvate (e.g., hydrate), or solvate of a salt thereof, or a mixture thereof.
  • the solid form is an amorphous form.
  • the solid form is substantially pure.
  • the solid form is substantially chemically pure.
  • the solid form is substantially physically pure.
  • the solid form has a chemical and/or physical purity of at least about 95% w/w. In one embodiment, the solid form has a chemical and/or physical purity of at least about 96% w/w. In one embodiment, the solid form has a chemical and/or physical purity of at least about 97% w/w. In one embodiment, the solid form has a chemical and/or physical purity of about 99.71% w/w. In one embodiment, the solid form has a chemical and/or physical purity of about 99.9% w/w. In one embodiment, the solid form is substantially enantiomerically pure. In one embodiment, the solid form (e.g. Form 1) has an enantiomeric purity of at least about 98% (e.g. 99% or 99.5%).
  • the solid form (e.g. Form 1) has an enantiomeric purity of at least about 98.0%. In one embodiment, the solid form (e.g. Form 1) has an enantiomeric purity of at least about 98.5%. In one embodiment, the solid form (e.g. Form 1) has an enantiomeric purity of at least about 99.0%. In one embodiment, the solid form (e.g. Form 1) has an enantiomeric purity of at least about 99.5%. In one embodiment, the solid form (e.g. Form 1) has an enantiomeric purity of about 99.9%. In one embodiment, the solid form (e.g. Form 1) has an enantiomeric purity of about 100%.
  • the compound of formula (I) is described in international patent application No. PCT/US2021/030842, the entirety of which is incorporated herein by reference.
  • provided herein is a solid form comprising a free base of Compound 1. In one embodiment, provided herein is a solid form comprising an anhydrous free base of Compound 1. In one embodiment, provided herein is a solid form comprising a solvate of a free base of Compound 1. In one embodiment, provided herein is a solid form comprising a hydrate of a free base of Compound 1. In one embodiment, provided herein is a solid form comprising a MTBE, MEK, ACN, or 1,4-dioxane solvate of a free base of Compound 1.
  • Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof can exist in a variety of solid forms.
  • Such solid forms include crystalline solids (e.g., crystalline forms of anhydrous Compound 1, crystalline forms of hydrates of Compound 1, and crystalline forms of solvates of Compound 1), amorphous solids, or mixtures of crystalline and amorphous solids.
  • the solid form is substantially crystalline.
  • the solid form is crystalline.
  • the molar ratio of Compound 1 to the solvent (e.g. water) in the solid form ranges from about 10:1 to about 1:10. In some embodiments, the molar ratio of Compound 1 to the solvent (e.g. water) in the solid form ranges from about 5:1 to about 1:5. In some embodiments, the molar ratio of Compound 1 to the solvent (e.g. water) in the solid form ranges from about 3:1 to about 1:3. In some embodiments, the molar ratio of Compound 1 to the solvent (e.g. water) in the solid form ranges from about 2:1 to about 1:2. In one embodiment, the molar ratio is about 1:2 (i.e., bis-solvate or dihydrate). In another embodiment, the molar ratio is about 1:1 (i.e., mono-solvate or mono-hydrate). In yet another embodiment, the molar ratio is about 2:1 (i.e., hemi-solvate or hemi-hydrate).
  • a Form 1 of Compound 1 in one embodiment, provided herein is a Form 1 of Compound 1.
  • a representative XRPD pattern of Form 1 of Compound 1 is provided in FIG. 1 .
  • a solid form comprising a free base of Compound 1, characterized by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22 or all of the XRPD peaks located at approximately the following positions (e.g., degrees 2 ⁇ 0.2) when measured using Cu K ⁇ radiation: 10.7, 12.0, 12.2, 13.9, 15.0, 17.4, 18.4, 18.6, 20.8, 21.2, 21.3, 21.6, 21.7, 23.3, 23.5, 24.0, 25.2, 26.0, 26.2, 26.7, 27.7, 28.0, and 29.6° 2 ⁇ .
  • the solid form is characterized by 3 of the peaks.
  • the solid form is characterized by 5 of the peaks.
  • the solid form is characterized by 7 of the peaks. In one embodiment, the solid form is characterized by 9 of the peaks. In one embodiment, the solid form is characterized by 11 of the peaks. In one embodiment, the solid form is characterized by all of the peaks.
  • a solid form (e.g. a crystalline form) comprising a free base of Compound 1, characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 10.7, 15.0, 17.4, 20.8, 21.2, 21.3, 21.6, 24.0, and 25.2° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 10.7, 15.0, 17.4, 20.8, 21.2, 21.3, 21.6, 24.0, and 25.2° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 10.7, 15.0, 17.4, 20.8, 21.2, 21.3, 21.6, 24.0, and 25.2° 2 ⁇ .
  • a solid form e.g. a crystalline form
  • a free base of Compound 1 characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 10.7, 12.0, 12.2, 13.9, 15.0, 17.4, 18.4, 20.8, 21.2, 21.3, 21.6, 24.0, and 25.2° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 10.7, 12.0, 12.2, 13.9, 15.0, 17.4, 18.4, 20.8, 21.2, 21.3, 21.6, 24.0, and 25.2° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 10.7, 12.0, 12.2, 13.9, 15.0, 17.4, 18.4, 20.8, 21.2, 21.3, 21.6, 24.0, and 25.2° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern comprising peaks at approximately (e.g., ⁇ 0.2°) 10.7, 15.0, and 21.2° 2 ⁇ .
  • the XRPD pattern further comprises peaks at approximately (e.g., 0.2°) 17.4 and 21.3° 2 ⁇ .
  • the XRPD pattern further comprises peaks at approximately (e.g., 0.2°) 12.0, 12.2, and 13.9° 2 ⁇ .
  • the XRPD pattern further comprises peaks at approximately (e.g., ⁇ 0.2°) 21.6 and 24.0° 2 ⁇ .
  • the XRPD pattern comprises peaks at approximately (e.g., 0.2°) 10.7, 15.0, 17.4, 20.8, 21.2, 21.3, 21.6, 24.0 and 25.2° 2 ⁇ . In one embodiment, the XRPD pattern comprises peaks at approximately (e.g., ⁇ 0.2°) 10.7, 12.0, 12.2, 13.9, 15.0, 17.4, 18.4, 20.8, 21.2, 21.3, 21.6, 24.0 and 25.2° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern that matches the XRPD pattern depicted in FIG. 1 .
  • an XRPD pattern described herein is obtained using Cu K ⁇ radiation.
  • the XRPD pattern is measured by XRPD using Cu K ⁇ radiation comprising K ⁇ 1 radiation having a wavelength of 1.5406 ⁇ and K ⁇ 2 radiation having a wavelength of 1.5444 ⁇ , wherein the K ⁇ 1 :K ⁇ 2 ratio is 0.5.
  • a representative DSC thermogram of Form 1 is provided in FIG. 3 .
  • a solid form comprising a free base of Compound 1, which exhibits, as characterized by DSC, a thermal event (endothermic) with an onset temperature of about 265° C. (e.g. ⁇ 2°).
  • the thermal event also has a peak temperature of about 267° C. (e.g. ⁇ 2°).
  • the thermal event corresponds to melting.
  • the solid form is characterized by a DSC thermogram that matches the DSC thermogram depicted in FIG. 3 .
  • the DSC thermogram is as measured by DSC using a scanning rate of about 10° C./minute.
  • the solid form has a melting point at about 270° C. (e.g. ⁇ 2°).
  • a representative overlay of TGA/DSC thermograms of Form 1 is provided in FIG. 2 .
  • a solid form comprising a free base of Compound 1, which exhibits substantially no weight loss upon heating from about 20° C. to about 200° C.
  • a solid form comprising a free base of Compound 1, which exhibits decomposition upon heating from about 200° C. (e.g. ⁇ 2°).
  • the solid form is characterized by a TGA thermogram that matches the TGA thermogram depicted in FIG. 2 .
  • the TGA thermogram is as measured using a heating rate of about 10° C./minute.
  • a representative DVS isotherm of Form 1 is provided in FIG. 4 .
  • a solid form comprising a free base of Compound 1, which exhibits a weight increase of about 0.25% (e.g. ⁇ 0.05%) when subjected to an increase in relative humidity from about 0 to about 90% relative humidity.
  • the solid form is characterized by a DVS isotherm that matches the DVS isotherm depicted in FIG. 4 .
  • the DVS isotherm is as measured at about 25° C.
  • a representative FT-IR spectrum of Form 1 is provided in FIG. 5 .
  • a solid form comprising a free base of Compound 1, characterized by an FT-IR spectrum comprising resonances at approximately 1621, 3523, 3413 (e.g. ⁇ 5) cm-1.
  • the FT-IR spectrum comprises resonances at approximately at least three peaks selected from the group consisting of 1176, 1400, 1537, 1621, 3413, and 3523 (e.g. ⁇ 5) cm-1.
  • a solid form comprising a free base of Compound 1, characterized by an FT-IR spectrum that matches the FT-IR spectrum depicted in FIG. 5 .
  • Form 1 has a unit cell of a space group of P2 1 .
  • Form 1 has a volume of about 1022.7 ⁇ 3 /cell.
  • Form 1 has a Z value of 2.
  • Form 1 has a density of about 1.362 g/cm 3 .
  • a solid form comprising a free base of Compound 1 which is anhydrous.
  • the solid form is a crystalline anhydrous free base of Compound 1.
  • the solid form is substantially free of amorphous Compound 1.
  • the solid form is substantially free of other crystalline forms of Compound 1.
  • the solid form is substantially free of salts of Compound 1.
  • the solid form is not solvated.
  • one or more residual solvent e.g., small amount of ethyl acetate
  • the residual solvent may be present in the solid form, but the residual solvent does not form a solvate of Compound 1.
  • the solid form is substantially pure.
  • the solid form is substantially chemically pure.
  • the solid form is substantially physically pure. In one embodiment, the solid form has a chemical and/or physical purity of at least about 97% w/w. In one embodiment, the solid form has a chemical and/or physical purity of at least about 98% w/w. In one embodiment, the solid form has a chemical and/or physical purity of at least about 99% w/w. In one embodiment, the solid form has a chemical and/or physical purity of about about 99.71% w/w. In one embodiment, the solid form has a chemical and/or physical purity of about 99.9% w/w. In one embodiment, the solid form has an enantiomeric purity of at least about 98% (e.g. 99% or 99.5%).
  • the solid form has an enantiomeric purity of at least about about 98.5%. In one embodiment, the solid form has an enantiomeric purity of at least about about 99.0%. In one embodiment, the solid form has an enantiomeric purity of at least about about 99.5%. In one embodiment, the solid form has an enantiomeric purity of about 99.9%. In one embodiment, the solid form (e.g. Form 1) has an enantiomeric purity of about 100%. In one embodiment, the purity is determined by HPLC or chiral HPLC (% area).
  • the solid form is non-hygroscopic. In one embodiment, the solid form is stable after storage at 40° C./75% RH, 25° C./60% RH and/or 80° C. for at least 7 days. In embodiment, the solid form is stable to compression at about 100 MPa and/or about 250 MPa for at least about 60 seconds.
  • provided herein is a solid form comprising Form 1 of a free base of Compound 1 and amorphous free base of Compound 1. In one embodiment, provided herein is a solid form comprising Form 1 of a free base Compound 1 and one or more other crystalline forms of a free base of Compound 1 provided herein. In one embodiment, provided herein is a solid form comprising Form 1 of a free base Compound 1 and Form 7 of a free base of Compound 1 provided herein.
  • a Form 2 of Compound 1 is provided herein.
  • a representative XRPD pattern of Form 2 of Compound 1 is provided in FIG. 7 .
  • a solid form comprising a free base of Compound 1, characterized by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or all of the XRPD peaks located at approximately the following positions (e.g., degrees 2 ⁇ 0.2) when measured using Cu K ⁇ radiation: 6.8, 8.6, 10.0, 10.7, 13.8, 14.0, 14.9, 15.9, 17.3, 18.3, 18.9, 19.4, 20.1, 20.4, 20.8, 21.2, 21.7, 22.9, 23.3, 23.8, 24.2, 24.8, 25.1, and 30.4° 2 ⁇ .
  • the solid form is characterized by 3 of the peaks.
  • the solid form is characterized by 5 of the peaks.
  • the solid form is characterized by 7 of the peaks. In one embodiment, the solid form is characterized by 9 of the peaks. In one embodiment, the solid form is characterized by 11 of the peaks. In one embodiment, the solid form is characterized by all of the peaks.
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 6.8, 8.6, 10.0, 13.8, 14.0, 20.1, 20.8, 21.2, and 22.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 6.8, 8.6, 10.0, 13.8, 14.0, 20.1, 20.8, 21.2, and 22.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 6.8, 8.6, 10.0, 13.8, 14.0, 20.1, 20.8, 21.2, and 22.9° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern comprising peaks at approximately (e.g., ⁇ 0.2°) 8.6, 14.0, and 20.8° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., 0.2°) 6.8 and 13.8° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 10.0 and 21.2° 2 ⁇ .
  • the XRPD pattern comprises peaks at approximately (e.g., ⁇ 0.2°) 6.8, 8.6, 10.0, 13.8, 14.0, 20.1, 20.8, 21.2 and 22.9° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern that matches the XRPD pattern depicted in FIG. 7 .
  • an XRPD pattern described herein is obtained using Cu K ⁇ radiation.
  • the XRPD pattern is measured by XRPD using Cu K ⁇ radiation comprising K ⁇ 1 radiation having a wavelength of 1.5406 ⁇ and K ⁇ 2 radiation having a wavelength of 1.5444 ⁇ , wherein the K ⁇ 1 :K ⁇ 2 ratio is 0.5.
  • a representative overlay of TGA/DTA thermograms of Form 2 is provided in FIG. 8 .
  • a solid form comprising a free base of Compound 1, which exhibits, as characterized by DTA, a thermal event (endo) with an onset temperature of about 98° C. (e.g. ⁇ 2°).
  • the solid form is characterized by a DTA thermogram that matchs the DTA thermogram depicted in FIG. 8 .
  • a solid form comprising a free base of Compound 1, which exhibits a weight loss of about 9.8% upon heating from about 90° C. to about 130° C.
  • the solid form is characterized by a TGA thermogram that matches the TGA thermogram depicted in FIG. 8 .
  • FIG. 9 Another representative overlay of TGA/DTA thermograms of Form 2 is provided in FIG. 9 .
  • a solid form comprising a free base of Compound 1, which exhibits, as characterized by DTA, a thermal event (endo) with an onset temperature of about 96° C. (e.g. ⁇ 2°) and/or a thermal event (exo) with an onset temperature of about 169° C. (e.g. ⁇ 2°).
  • the solid form is characterized by a DTA thermogram is substantially in accordance with that depicted in FIG. 9 .
  • a solid form which exhibits a weight loss of about 15.4% upon heating from about 50° C. to about 170° C.
  • the solid form is characterized by a TGA thermogram that matches the TGA thermogram depicted in FIG. 9 .
  • the DTA thermogram is as measured by DTA using a scanning rate of about 10° C./minute. In one embodiment, the TGA thermogram is as measured using a heating rate of about 10° C./minute.
  • a representative DSC thermograms of Form 2 is provided in FIG. 10 .
  • a solid form comprising a free base of Compound 1, which exhibits, as characterized by DSC, a thermal event (endo) with an onset temperature of about 96° C. (e.g. ⁇ 2°). In one embodiment, the thermal event also has a peak temperature of about 99° C. (e.g. ⁇ 2°).
  • a solid form comprising a free base of Compound 1, which exhibits, as characterized by DSC, a thermal event (exo) with an onset temperature of about 168° C. (e.g. ⁇ 2°).
  • the thermal event also has a peak temperature of about 174° C. (e.g. ⁇ 2°).
  • the solid form is characterized by a DSC thermogram that matches the DSC thermogram depicted in FIG. 10 .
  • the DSC thermogram is as measured by DSC using a scanning rate of about 10° C./minute.
  • a representative FT-IR spectrum of Form 2 is provided in FIG. 11 .
  • a solid form comprising a free base of Compound 1, characterized by an FT-IR spectrum that matches the FT-IR spectrum depicted in FIG. 11 .
  • a solid form comprising a free base of Compound 1 which is a crystalline solvate of free base of Compound 1.
  • the solid form is substantially free of amorphous Compound 1.
  • the solid form is substantially free of other solid forms (e.g., crystalline forms) of Compound 1.
  • the solid form is substantially free of salts of Compound 1.
  • the solid form is provided as substantially pure.
  • the solid form is substantially chemically pure.
  • the solid form is substantially physically pure.
  • the solid form is non-hygroscopic.
  • the solid form is stable after storage at 40° C./75% RH or 25° C./60% RH for at least 7 days. In one embodiment, the solid form undergoes a form change upon heating. In one embodiment, the solid form undergoes a form change after being stored at about 80° C. In one embodiment, the form change results in Form 7 of a free base of Compound 1.
  • a solid form comprising a free base of Compound 1, wherein the molar ratio of Compound 1 to the solvent ranges from about 1:0.5 to about 1:2. In one embodiment, the molar ratio of Compound 1 to the solvent ranges from about 1:0.6 to about 1:1.35. In one embodiment, the molar ratio of Compound 1 to the solvent ranges from about 1:0.75 to about 1:1. In one embodiment, the molar ratio of Compound 1 to the solvent is about 1:0.6. In one embodiment, the molar ratio of Compound 1 to the solvent is about 1:0.9. In one embodiment, the molar ratio of Compound 1 to the solvent is about 1:1.3. In one embodiment, the molar ratio of Compound 1 to the solvent is about 1:1.
  • a solid form comprising a free base of Compound 1 which is an isostructural solvate.
  • a solid form comprising a free base of Compound 1 which is a methyl ethyl ketone (MEK) solvate of free base of Compound 1.
  • MEK methyl ethyl ketone
  • the molar ratio of Compound 1 to MEK is about 1:0.6.
  • the solid form is a methyl t-butyl ether solvate of free base of Compound 1.
  • the molar ratio of Compound 1 to methyl t-butyl ether is about 1:0.9.
  • the molar ratio of Compound 1 to methyl t-butyl ether is about 1:1.3.
  • the solid form is a 1-butanol, 2-methyl-1-propanol, 3-methyl-1-butanol, acetone, cyclopentyl methyl ether, or tAmyl alcohol solvate of free base of Compound 1.
  • provided herein is a solid form comprising Form 2 of a free base of Compound 1 and amorphous free base of Compound 1. In one embodiment, provided herein is a solid form comprising Form 2 of a free base Compound 1 and one or more other crystalline forms of a free base of Compound 1 provided herein.
  • a Form 3 of Compound 1 is provided herein.
  • a representative XRPD pattern of Form 3 of Compound 1 is provided in FIG. 12 .
  • a solid form comprising a free base of Compound 1, characterized by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 or all of the XRPD peaks located at approximately the following positions (e.g., degrees 2 ⁇ 0.2) when measured using Cu K ⁇ radiation: 9.7, 11.4, 11.8, 12.5, 13.4, 14.7, 16.2, 16.9, 18.9, 19.3, 19.5, 20.9, 21.9, 22.5, 22.9, 23.3, and 24.3° 2 ⁇ .
  • the solid form is characterized by 3 of the peaks.
  • the solid form is characterized by 5 of the peaks.
  • the solid form is characterized by 7 of the peaks.
  • the solid form is characterized by 9 of the peaks.
  • the solid form is characterized by 11 of the peaks.
  • the solid form is characterized by all of the peaks.
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 9.7, 11.4, 13.4, 16.2, 18.9, 19.5, 20.9, 22.5, 22.9, and 23.3° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 9.7, 11.4, 13.4, 16.2, 18.9, 19.5, 20.9, 22.5, 22.9, and 23.3° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 9.7, 11.4, 13.4, 16.2, 18.9, 19.5, 20.9, 22.5, 22.9, and 23.3° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern comprising peaks at approximately (e.g., ⁇ 0.2°) 13.4, 19.5, and 20.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., 0.2°) 11.4 and 22.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 9.7 and 16.2° 2 ⁇ .
  • the XRPD pattern comprises peaks at approximately (e.g., ⁇ 0.2°) 9.7, 11.4, 13.4, 16.2, 18.9, 19.5, 20.9, 22.5, 22.9, and 23.3° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern that matches the XRPD pattern depicted in FIG. 12 .
  • an XRPD pattern described herein is obtained using Cu K ⁇ radiation.
  • the XRPD pattern is measured by XRPD using Cu K ⁇ radiation comprising K ⁇ 1 radiation having a wavelength of 1.5406 ⁇ and K ⁇ 2 radiation having a wavelength of 1.5444 ⁇ , wherein the K ⁇ 1 :K ⁇ 2 ratio is 0.5.
  • a representative overlay of TGA/DTA thermograms of Form 3 is provided in FIG. 13 .
  • a solid form comprising a free base of Compound 1, which exhibits, as characterized by DTA, a thermal event with an onset temperature of about 168° C. (e.g. ⁇ 2°).
  • the solid form is characterized by a DTA thermogram that matches the DTA thermogram depicted in FIG. 13 .
  • the DTA thermogram is as measured by DTA using a scanning rate of about 10° C./minute.
  • a solid form comprising a free base of Compound 1, which exhibits a weight loss of about 1.3% upon heating from about 100° C. to about 130° C.
  • the solid form is characterized by a TGA thermogram that matches the TGA thermogram depicted in FIG. 13 .
  • the TGA thermogram is as measured using a heating rate of about 10° C./minute.
  • a solid form comprising a free base of Compound 1, which is a crystalline solvate of free base of Compound 1.
  • the solid form is substantially free of amorphous Compound 1.
  • the solid form is substantially free of other solid forms (e.g., crystalline forms) of Compound 1.
  • the solid form is substantially free of salts of Compound 1.
  • the solid form is provided as substantially pure.
  • the solid form is substantially chemically pure.
  • the solid form is substantially physically pure.
  • a solid form comprising a free base of Compound 1 which is an isostructural solvate.
  • a solid form comprising a free base of Compound 1, wherein the molar ratio of Compound 1 to the solvent ranges from about 1:0.2 to about 1:1.
  • the solid form is a hydrate of free base of Compound 1.
  • the solid form is a hemihydrate of free base of Compound 1.
  • the solid form is a methanol solvate of a free base of Compound 1.
  • the solid form is an isopropyl acetate solvate of a free base of Compound 1.
  • provided herein is a solid form comprising Form 3 of a free base of Compound 1 and amorphous free base of Compound 1. In one embodiment, provided herein is a solid form comprising Form 3 of a free base Compound 1 and one or more other crystalline forms of a free base of Compound 1 provided herein.
  • a Form 4 of Compound 1 is provided herein.
  • a representative XRPD pattern of Form 4 of Compound 1 is provided in FIG. 14 .
  • a solid form comprising a free base of Compound 1, characterized by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or all of the XRPD peaks located at approximately the following positions (e.g., degrees 2 ⁇ 0.2) when measured using Cu K ⁇ radiation: 7.5, 7.6, 10.8, 12.0, 12.1, 12.7, 12.8, 15.0, 15.7, 18.4, 19.0, 19.3, 19.7, 20.0, 21.4, 21.8, 21.9, 22.0, 22.8, 23.9, 24.1, 24.3, 24.8, 25.1, 25.7, 29.1, and 30.2° 2 ⁇ .
  • the solid form is characterized by 3 of the peaks.
  • the solid form is characterized by 5 of the peaks. In one embodiment, the solid form is characterized by 7 of the peaks. In one embodiment, the solid form is characterized by 9 of the peaks. In one embodiment, solid form is characterized by 11 of the peaks. In one embodiment, solid form is characterized by all of the peaks.
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 12.0, 12.1, 12.7, 12.8, 15.0, 15.7, 18.4, 21.4, 21.9, 23.9 and 24.3° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 12.0, 12.1, 12.7, 12.8, 15.0, 15.7, 18.4, 21.4, 21.9, 23.9 and 24.3° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 12.0, 12.1, 12.7, 12.8, 15.0, 15.7, 18.4, 21.4, 21.9, 23.9 and 24.3° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern comprising peaks at approximately (e.g., ⁇ 0.2°) 12.1, 12.7, and 18.4° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., 0.2°) 15.0 and 21.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 23.9 and 24.3° 2 ⁇ .
  • the XRPD pattern comprises peaks at approximately (e.g., ⁇ 0.2°) 12.1, 12.7, 15.0, 15.7, 18.4, 21.4, 21.9, 23.9 and 24.3° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern that matches the XRPD pattern depicted in FIG. 14 .
  • an XRPD pattern described herein is obtained using Cu K ⁇ radiation.
  • the XRPD pattern is measured by XRPD using Cu K ⁇ radiation comprising K ⁇ 1 radiation having a wavelength of 1.5406 ⁇ and K ⁇ 2 radiation having a wavelength of 1.5444 ⁇ , wherein the K ⁇ 1 :K ⁇ 2 ratio is 0.5.
  • a representative overlay of TGA/DTA thermograms of Form 4 is provided in FIG. 15 .
  • a solid form comprising a free base of Compound 1, which exhibits, as characterized by DTA, a thermal event (endo) with an onset temperature of about 135° C. (e.g. ⁇ 2°).
  • the solid form is characterized by a DTA thermogram that matches the DTA therogram depicted in FIG. 15 .
  • the DTA thermogram is as measured by DTA using a scanning rate of about 10° C./minute.
  • a solid form comprising a free base of Compound 1, which exhibits a weight loss of about 9.0% upon heating from about 110° C. to about 145° C.
  • the solid form is characterized by a TGA thermogram that matches the TGA thermogram depicted in FIG. 15 .
  • the TGA thermogram is as measured using a heating rate of about 10° C./minute.
  • a solid form comprising a free base of Compound 1 which is a crystalline solvate of free base of Compound 1.
  • the solid form is substantially free of amorphous Compound 1.
  • the solid form is substantially free of other solid forms (e.g., crystalline forms) of Compound 1.
  • the solid form is substantially free of salts of Compound 1.
  • the solid form is provided as substantially pure.
  • the solid form is substantially chemically pure.
  • the solid form is substantially physically pure.
  • a solid form comprising a free base of Compound 1, wherein the molar ratio of Compound 1 to the solvent ranges from about 1:0.5 to about 1:1.5. In one embodiment, the molar ratio of Compound 1 to the solvent ranges from about 1:0.8 to about 1:1.1. In one embodiment, the solid form is an acetonitrile solvate of free base of Compound 1. In one embodiment, the molar ratio of Compound 1 to acetonitrile is about 1:0.9.
  • provided herein is a solid form comprising Form 4 of a free base of Compound 1 and amorphous free base of Compound 1. In one embodiment, provided herein is a solid form comprising Form 4 of a free base Compound 1 and one or more other crystalline forms of a free base of Compound 1 provided herein.
  • a Form 5 of Compound 1 is provided herein.
  • a representative XRPD pattern of Form 4 of Compound 1 is provided in FIG. 16 .
  • a solid form comprising a free base of Compound 1, characterized by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, or all of the XRPD peaks located at approximately the following positions (e.g., degrees 2 ⁇ 0.2) when measured using Cu K ⁇ radiation: 7.6, 10.5, 10.8, 12.1, 12.8, 13.4, 15.0, 15.8, 16.4, 18.3, 18.9, 19.3, 19.7, 20.3, 21.9, 22.7, 23.6, 23.9, and 28.9° 2 ⁇ .
  • the solid form is characterized by 3 of the peaks.
  • the solid form is characterized by 5 of the peaks.
  • the solid form is characterized by 7 of the peaks.
  • the solid form is characterized by 9 of the peaks.
  • the solid form is characterized by 11 of the peaks.
  • the solid form is characterized by all of the peaks.
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 7.6, 10.5, 10.8, 12.1, 12.8, 15.0, 15.8, 19.7, 21.9, 22.7, and 23.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 7.6, 10.5, 10.8, 12.1, 12.8, 15.0, 15.8, 19.7, 21.9, 22.7, and 23.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 7.6, 10.5, 10.8, 12.1, 12.8, 15.0, 15.8, 19.7, 21.9, 22.7, and 23.9° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern comprising peaks at approximately (e.g., ⁇ 0.2°) 10.5, 10.8, and 21.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., 0.2°) 12.1 and 12.8° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 7.6 and 15.0° 2 ⁇ .
  • the XRPD pattern comprises peaks at approximately (e.g., ⁇ 0.2°) 7.6, 10.5, 10.8, 12.1, 12.8, 15.0, 15.8, 19.7 and 21.9° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern that matches the XRPD pattern depicted in FIG. 16 .
  • an XRPD pattern described herein is obtained using Cu K ⁇ radiation.
  • the XRPD pattern is measured by XRPD using Cu K ⁇ radiation comprising K ⁇ 1 radiation having a wavelength of 1.5406 ⁇ and K ⁇ 2 radiation having a wavelength of 1.5444 ⁇ , wherein the K ⁇ 1 :K ⁇ 2 ratio is 0.5.
  • a solid form comprising a free base of Compound 1 which is a crystalline solvate of free base of Compound 1.
  • the solid form is substantially free of amorphous Compound 1.
  • the solid form is substantially free of other solid forms (e.g., crystalline forms) of Compound 1.
  • the solid form is substantially free of salts of Compound 1.
  • the solid form is provided as substantially pure.
  • the solid form is substantially chemically pure.
  • the solid form is substantially physically pure.
  • a solid form comprising a free base of Compound 1, wherein the molar ratio of Compound 1 to the solvent ranges from about 1:0.5 to about 1:1.5. In one embodiment, the molar ratio of Compound 1 to the solvent ranges from about 1:0.6 to about 1:0.8. In one embodiment, the solid form is an acetonitrile solvate of free base of Compound 1. In one embodiment, the molar ratio of Compound 1 to acetonitrile is about 1:0.7.
  • provided herein is a solid form comprising Form 5 of a free base of Compound 1 and amorphous free base of Compound 1. In one embodiment, provided herein is a solid form comprising Form 5 of a free base Compound 1 and one or more other crystalline forms of a free base of Compound 1 provided herein.
  • a Form 6 of Compound 1 is provided herein.
  • a representative XRPD pattern of Form 6 of Compound 1 is provided in FIG. 17 .
  • a solid form comprising a free base of Compound 1, characterized by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or all of the XRPD peaks located at approximately the following positions (e.g., degrees 2 ⁇ 0.2) when measured using Cu K ⁇ radiation: 6.7, 6.8, 8.3, 8.5, 8.6, 10.0, 13.8, 13.9, 14.1, 15.9, 17.3, 18.3, 18.7, 18.9, 19.3, 20.1, 20.5, 20.8, 21.2, 22.9, 23.9, 24.2, 24.8, 25.2, and 26.8° 2 ⁇ .
  • the solid form is characterized by 3 of the peaks.
  • the solid form is characterized by 5 of the peaks.
  • the solid form is characterized by 7 of the peaks. In one embodiment, the solid form is characterized by 9 of the peaks. In one embodiment, the solid form is characterized by 11 of the peaks. In one embodiment, the solid form is characterized by all of the peaks.
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 8.4, 8.6, 10.0, 15.9, 18.7, 20.5, 20.8, 21.2 and 22.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 8.4, 8.6, 10.0, 15.9, 18.7, 20.5, 20.8, 21.2 and 22.9° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 8.4, 8.6, 10.0, 15.9, 18.7, 20.5, 20.8, 21.2 and 22.9° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern comprising peaks at approximately (e.g., ⁇ 0.2°) 8.6, 18.7, and 20.5° 2 ⁇ .
  • the solid from is characterized by an XRPD pattern further comprising peaks at approximately (e.g., 0.2°) 15.9 and 20.8° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 10.0 and 14.1° 2 ⁇ .
  • the XRPD pattern comprises peaks at approximately (e.g., ⁇ 0.2°) 8.6, 10.0, 15.9, 18.7, 20.5, 20.8, 21.2 and 22.9° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern that matches the XRPD pattern depicted in FIG. 17 .
  • an XRPD pattern described herein is obtained using Cu K ⁇ radiation.
  • the XRPD pattern is measured by XRPD using Cu K ⁇ radiation comprising K ⁇ 1 radiation having a wavelength of 1.5406 ⁇ and K ⁇ 2 radiation having a wavelength of 1.5444 ⁇ , wherein the K ⁇ 1 :K ⁇ 2 ratio is 0.5.
  • a representative overlay of TGA/DTA thermograms of Form 6 is provided in FIG. 18 .
  • a solid form comprising a free base of Compound 1, which exhibits, as characterized by DTA, a thermal event (endo) with an onset temperature of about 126° C. (e.g. ⁇ 2°).
  • the solid form is characterized by a DTA thermogram that matches the DTA thermogram depicted in FIG. 18 .
  • the DTA thermogram is as measured by DTA using a scanning rate of about 10° C./minute.
  • a solid form comprising a free base of Compound 1, which exhibits a weight loss of about 13.1% upon heating from about 25° C. to about 50° C. and/or a weight loss of about 12.1% upon heating from about 115° C. to about 155° C.
  • the solid form is characterized by a TGA thermogram that matches the TGA thermogram depicted in FIG. 18 .
  • the TGA thermogram is as measured using a heating rate of about 10° C./minute.
  • a solid form comprising a free base of Compound 1 which is a crystalline solvate of free base of Compound 1.
  • the solid form is substantially free of amorphous Compound 1.
  • the solid form is substantially free of other solid forms (e.g., crystalline forms) of Compound 1.
  • the solid form is substantially free of salts of Compound 1.
  • the solid form is provided as substantially pure.
  • the solid form is substantially chemically pure.
  • the solid form is substantially physically pure.
  • a solid form comprising a free base of Compound 1, wherein the molar ratio of Compound 1 to the solvent ranges from about 1:0.2 to about 1:1.5. In one embodiment, the molar ratio of Compound 1 to the solvent ranges from about 1:0.5 to about 1:1.3. In one embodiment, the solid form is a 1,4-dioxane solvate of free base of Compound 1. In one embodiment, the molar ratio of Compound 1 to 1,4-dioxane is about 1:0.7. In another embodiment, the molar ratio of Compound 1 to 1,4-dioxane is about 1:1.2.
  • provided herein is a solid form comprising Form 6 of a free base of Compound 1 and amorphous free base of Compound 1. In one embodiment, provided herein is a solid form comprising Form 6 of a free base Compound 1 and one or more other crystalline forms of a free base of Compound 1 provided herein.
  • a Form 7 of Compound 1 is provided herein.
  • a representative XRPD pattern of Form 7 of Compound 1 is provided in FIG. 19 .
  • a solid form comprising a free base of Compound 1, characterized by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 or all of the XRPD peaks located at approximately the following positions (e.g., degrees 2 ⁇ 0.2) when measured using Cu K ⁇ radiation: 9.5, 9.7, 11.1, 11.4, 12.3, 12.5, 13.4, 14.6, 14.7, 15.8, 16.2, 16.9, 18.4, 18.8, 19.3, 19.5, 20.9, 21.5, 22.5, 22.8, 23.2, 24.9, 25.1, and 29.5° 2 ⁇ .
  • the solid form is characterized by 3 of the peaks.
  • the solid form is characterized by 5 of the peaks.
  • the solid form is characterized by 7 of the peaks. In one embodiment, the solid form is characterized by 9 of the peaks. In one embodiment, the solid form is characterized by 11 of the peaks. In one embodiment, the solid form is characterized by all of the peaks.
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 9.5, 9.7, 11.4, 12.3, 12.5, 13.4, 14.6, 14.7, 15.8, 20.9, 22.8 and 23.2° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 9.5, 9.7, 11.4, 12.3, 12.5, 13.4, 14.6, 14.7, 15.8, 20.9, 22.8 and 23.2° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 9.5, 9.7, 11.4, 12.3, 12.5, 13.4, 14.6, 14.7, 15.8, 20.9, 22.8 and 23.2° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern comprising peaks at approximately (e.g., ⁇ 0.2°) 12.5, 13.4, and 14.6° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., 0.2°) 20.9 and 22.8° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 11.4 and 15.8° 2 ⁇ .
  • the XRPD pattern comprises peaks at approximately (e.g., ⁇ 0.2°) 9.5, 11.4, 12.5, 13.4, 14.6, 15.8, 20.9, 22.8 and 23.2° 2 ⁇ .
  • a solid form comprising a free base of Compound 1, characterized by an XRPD pattern that matches the XRPD pattern depicted in FIG. 19 .
  • an XRPD pattern described herein is obtained using Cu K ⁇ radiation.
  • the XRPD pattern is measured by XRPD using Cu K ⁇ radiation comprising K ⁇ 1 radiation having a wavelength of 1.5406 ⁇ and K ⁇ 2 radiation having a wavelength of 1.5444 ⁇ , wherein the K ⁇ 1 :K ⁇ 2 ratio is 0.5.
  • a representative DSC thermograms of Form 7 is provided in FIG. 20 .
  • a solid form comprising a free base of Compound 1, which exhibits, as characterized by DSC, a thermal event (endo) with an onset temperature of about 278° C. (e.g. ⁇ 2°).
  • the thermal event also has a peak temperature of about 280° C. (e.g. ⁇ 2°).
  • the thermal event corresponds to melting.
  • the solid form is characterized by a DSC thermogram that matches the DSC thermogram depicted in FIG. 20 .
  • the DSC thermogram is as measured by DSC using a scanning rate of about 10° C./minute.
  • a representative DVS isotherm of Form 7 is provided in FIG. 21 .
  • a solid form comprising a free base of Compound 1, which exhibits a weight increase of about 0.27% (e.g. ⁇ 0.05%) when subjected to an increase in relative humidity from about 0 to about 90% relative humidity.
  • the solid form is characterized by a DVS isotherm that matches the DVS isotherm depicted in FIG. 21 .
  • the DVS isotherm is as measured at about 25° C.
  • a representative FT-IR spectrum of Form 7 is provided in FIG. 22 .
  • a solid form comprising a free base of Compound 1, characterized by an FT-IR spectrum comprising resonances at approximately 3386 and 3489 (e.g. ⁇ 5) cm-1.
  • the FT-IR spectrum comprises at least three resonances selected from the group approximately 1461, 1584, 1619, 3386 and 3489 (e.g. ⁇ 5) cm-1.
  • a solid form comprising a free base of Compound 1, characterized by an FT-IR spectrum that matches the FT-IR spectrum depicted in FIG. 22 .
  • the solid form has a unit cell of a space group of P2 1 2 1 2 1 .
  • the solid form has a volume of about 2124.1 ⁇ 3 /cell.
  • the solid form has a Z value of 4.
  • the solid form has a density of about 1.312 g/cm 3 .
  • a solid form comprising a free base of Compound 1 which is anhydrous.
  • the solid form is a crystalline anhydrous free base of Compound 1.
  • the solid form is substantially free of amorphous Compound 1.
  • the solid form is substantially free of other solid forms (i.e., crystalline forms) of Compound 1.
  • the solid form is substantially free of salts of Compound 1.
  • the solid form is provided as substantially pure.
  • one or more residual solvent e.g., small amount of ethyl acetate
  • the solid form is not solvated.
  • the solid form is substantially chemically pure.
  • the solid form is substantially physically pure.
  • provided herein is a solid form comprising Form 7 of a free base of Compound 1 and amorphous free base of Compound 1. In one embodiment, provided herein is a solid form comprising Form 7 of a free base Compound 1 and one or more other crystalline forms of a free base of Compound 1 provided herein.
  • the solvent is 2-MeTHF, isopropyl acetate, acetone, anisole, ethanol, ethyl acetate, isopropyl acetate, methyl ethyl ketone, or a mixture thereof.
  • the solvent is ethyl acetate.
  • the anti-solvent is water.
  • the anti-solvent is a non-polar organic solvent.
  • the non-polar organic solvent is a hydrocarbon solvent.
  • the anti-solvent is heptane.
  • the solvent is ethyl acetate and the anti-solvent is heptane.
  • the final ratio of solvent to anti-solvent is from about 1:2 to about 1:6. In one embodiment, the final ratio of solvent to anti-solvent is from about 1:3 to about 1:5. In one embodiment, the final ratio of solvent to anti-solvent is about 1:4. In one embodiment, the anti-solvent is added to the solvent at a temperature above room temperature. In one embodiment, the anti-solvent is added at a temperature of from about 30° C. to about 60° C. In one embodiment, the temperature is about 50° C.
  • the non-Form 1 solid form is exposed to one solvent. In one embodiment, the non-Form 1 solid form is exposed to a mixture of two solvents. In one embodiment, the non-Form 1 solid form is exposed to one or more solvents.
  • the solvent is an organic solvent. In one embodiment, the solvent is an alcohol. In one embodiment, the solvent is 1-butanol, 1-propanol, 2-methyl-1-propanol, 2-butanol, ethanol, or tAmyl alcohol.
  • the solvent is tetrahydrofuran, 2-methyltetrahydrofuran (2-MeTHF), butyl acetate, cyclopentyl methyl ether (CPME), ethyl acetate, isopropyl acetate, methyl ethyl ketone, toluene, methylisobutyl ketone or methyl acetate.
  • the solvent is ethyl acetate.
  • the solvent system comprises a mixture of two solvents. In one embodiment, the solvent system is a mixture of two solvents.
  • the mixture of two solvents is a mixture of 1-butanol and heptane, a mixture of 1-propanol and heptane, a mixture of 2-MeTHF and heptane, a mixture of 2-propanol and heptane, a mixture of ethanol and water, a mixture of acetone and water, a mixture of butyl acetate and heptane, a mixture of dimethylsulfoxide and water, a mixture of ethyl acetate and heptane, a mixture of isopropyl acetate and heptane, or a mixture of methyl ethyl ketone and heptane.
  • the mixture of two solvents is a mixture of 1-butanol and heptane. In one embodiment, the volume ratio of 1-butanol to heptane is from about 1:10 to about 1:5. In one embodiment, the volume ratio of 1-butanol to heptane is about 2:9. In one embodiment, the mixture of two solvents is a mixture of 1-propanol and heptane. In one embodiment, the volume ratio of 1-propanol to heptane is from about 1:5 to about 1:2. In one embodiment, the volume ratio of 1-propanol to heptane is about 1:4. In one embodiment, the mixture of two solvents is a mixture of 2-MeTHF and heptane.
  • the volume ratio of 2-MeTHF to heptane is from about 1:3 to about 1:3. In one embodiment, the volume ratio of 2-MeTHF to heptane is about 1:1. In one embodiment, the mixture of two solvents is a mixture of 2-propanol and heptane. In one embodiment, the volume ratio of 2-propanol to heptane is from about 1:5 to about 1:2. In one embodiment, the volume ratio of 2-propanol to heptane is about 1:3. In one embodiment, the mixture of two solvents is a mixture of 98.5% ethanol/1.5% water and water. In one embodiment, the volume ratio of 98.5% ethanol/1.5% water to water is from about 1:6 to about 1:2.
  • the volume ratio of 98.5% ethanol/1.5% water to water is about 2:5.
  • the mixture of two solvents is a mixture of acetone and water. In one embodiment, the volume ratio of acetone to water is from about 1:3 to about 2:1. In one embodiment, the volume ratio of acetone to water is about 1:1.
  • the mixture of two solvents is a mixture of butyl acetate and heptane. In one embodiment, the volume ratio of butyl acetate and heptane is from about 1:4 to about 2:1. In one embodiment, the volume ratio of butyl acetate and heptane is about 2:3.
  • the mixture of two solvents is a mixture of dimethylsulfoxide and water. In one embodiment, the volume ratio of dimethylsulfoxide and water is from about 1:1 to about 1:3. In one embodiment, the volume ratio of dimethylsulfoxide and water is about 1:2. In one embodiment, the mixture of two solvents is a mixture of ethanol and water. In one embodiment, the volume ratio of ethanol and water is from about 1:1 to about 1:3. In one embodiment, the volume ratio of ethanol and water is about 2:3. In one embodiment, the mixture of two solvents is a mixture of ethyl acetate and heptane. In one embodiment, the volume ratio of ethyl acetate and heptane is from about 1:1 to about 1:3.
  • the volume ratio of ethyl acetate and heptane is about 1:2. In one embodiment, the mixture of two solvents is a mixture of isopropyl acetate and heptane. In one embodiment, the volume ratio of isopropyl acetate and heptane is from about 1:1 to about 1:3. In one embodiment, the volume ratio of isopropyl acetate and heptane is about 2:3. In one embodiment, the mixture of two solvents is a mixture of methyl ethyl ketone and heptane. In one embodiment, the volume ratio of methyl ethyl ketone and heptane is from about 1:1 to about 1:3.
  • the volume ratio of methyl ethyl ketone and heptane is about 2:3.
  • the mixture of two solvents is a mixture of ethanol and heptane. In one embodiment, the volume ratio of ethanol and heptane is from about 1:10 to about 1:5. In one embodiment, the mixture of two solvents is a mixture of acetone and heptane. In one embodiment, the mixture of two solvents is a mixture of ethanol and heptane. In one embodiment, the volume ratio of acetone and heptane is from about 1:10 to about 1:5. In one embodiment, the mixture of two solvents is a mixture of tetrahydrofuran and heptane. In one embodiment, the mixture of two solvents is a mixture of ethanol and heptane. In one embodiment, the volume ratio of tetrahydrofuran and heptane is from about 1:10 to about 1:5.
  • the non-Form 1 solid form is an amorphous solid form of a compound of Formula (I). In one embodiment, the non-Form 1 solid form is any one of Form 2 to Form 7 of a compound of Formula (I). In one embodiment, the non-Form 1 solid form is Form 6. In one embodiment, the non-Form 1 solid form is Form 7.
  • the period of time sufficient to convert at least about 50% of the total amount of the non-Form 1 solid form into Form 1 is about 1 hr, about 2 hr, about 5 hr, about 10 hr, about 12 hr, about 20 hr, about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 1 of a compound of Formula (I) may be prepared by exposing a composition comprising a compound of Formula (I) to one or more solvent as described in the experiments provided herein, including but not limited to evaporation, anti-solvent addition, slow cooling, crash cooling, temperature cycling or solvent drop grinding.
  • Form 1 of a compound of Formula (I) is prepared by crystallization or recrystallization of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, from one or more solvents.
  • the solvent is an organic solvent.
  • the solvent is an alcohol.
  • the solvent is 1-butanol, 1-propanol, 2-methyl-1-propanol, 2-butanol, ethanol, or tAmyl alcohol.
  • the solvent is tetrahydrofuran, 2-methyltetrahydrofuran (2-MeTHF), butyl acetate, cyclopentyl methyl ether (CPME), ethyl acetate, isopropyl acetate, methyl ethyl ketone, toluene, or methyl acetate.
  • Form 1 of a compound of Formula (I) is prepared by crystallization or recrystallization of a compound of Formula (I) from a solvent comprising a mixture of two solvents.
  • the mixture of two solvents is a mixture of 1-butanol and heptane, a mixture of 1-propanol and heptane, a mixture of 2-MeTHF and heptane, a mixture of 2-propanol and heptane, a mixture of ethanol and water, a mixture of acetone and water, a mixture of butyl acetate and heptane, a mixture of dimethylsulfoxide and water, a mixture of ethyl acetate and heptane, a mixture of isopropyl acetate and heptane, or a mixture of methyl ethyl ketone and heptane.
  • the mixture of two solvents is a mixture of 1-butanol and heptane. In one embodiment, the volume ratio of 1-butanol to heptane is from about 1:10 to about 1:5. In one embodiment, the volume ratio of 1-butanol to heptane is about 2:9. In one embodiment, the mixture of two solvents is a mixture of 1-propanol and heptane. In one embodiment, the volume ratio of 1-propanol to heptane is from about 1:5 to about 1:2. In one embodiment, the volume ratio of 1-propanol to heptane is about 1:4. In one embodiment, the mixture of two solvents is a mixture of 2-MeTHF and heptane.
  • the volume ratio of 2-MeTHF to heptane is from about 1:3 to about 1:3. In one embodiment, the volume ratio of 2-MeTHF to heptane is about 1:1. In one embodiment, the mixture of two solvents is a mixture of 2-propanol and heptane. In one embodiment, the volume ratio of 2-propanol to heptane is from about 1:5 to about 1:2. In one embodiment, the volume ratio of 2-propanol to heptane is about 1:3. In one embodiment, the mixture of two solvents is a mixture of 98.5% ethanol/1.5% water and water. In one embodiment, the volume ratio of 98.5% ethanol/1.5% water to water is from about 1:6 to about 1:2.
  • the volume ratio of 98.5% ethanol/1.5% water to water is about 2:5.
  • the mixture of two solvents is a mixture of acetone and water. In one embodiment, the volume ratio of acetone to water is from about 1:3 to about 2:1. In one embodiment, the volume ratio of acetone to water is about 1:1.
  • the mixture of two solvents is a mixture of butyl acetate and heptane. In one embodiment, the volume ratio of butyl acetate and heptane is from about 1:4 to about 2:1. In one embodiment, the volume ratio of butyl acetate and heptane is about 2:3.
  • the mixture of two solvents is a mixture of dimethylsulfoxide and water. In one embodiment, the volume ratio of dimethylsulfoxide and water is from about 1:1 to about 1:3. In one embodiment, the volume ratio of dimethylsulfoxide and water is about 1:2. In one embodiment, the mixture of two solvents is a mixture of ethanol and water. In one embodiment, the volume ratio of ethanol and water is from about 1:1 to about 1:3. In one embodiment, the volume ratio of ethanol and water is about 2:3. In one embodiment, the mixture of two solvents is a mixture of ethyl acetate and heptane. In one embodiment, the volume ratio of ethyl acetate and heptane is from about 1:1 to about 1:3.
  • the volume ratio of ethyl acetate and heptane is about 1:2. In one embodiment, the mixture of two solvents is a mixture of isopropyl acetate and heptane. In one embodiment, the volume ratio of isopropyl acetate and heptane is from about 1:1 to about 1:3. In one embodiment, the volume ratio of isopropyl acetate and heptane is about 2:3. In one embodiment, the mixture of two solvents is a mixture of methyl ethyl ketone and heptane. In one embodiment, the volume ratio of methyl ethyl ketone and heptane is from about 1:1 to about 1:3. In one embodiment, the volume ratio of methyl ethyl ketone and heptane is about 2:3.
  • Form 1 of a compound of Formula (I) is prepared by crystallization or recrystallization as described in the experiments provided herein, including but not limited to evaporation, anti-solvent addition, slow cooling, or crash cooling.
  • Form 1 of a compound of Formula (I) is prepared by crystallization or recrystallization of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, from 2-MeTHF, isopropyl acetate, acetone, anisole, ethanol, ethyl acetate, isopropyl acetate, methyl ethyl ketone, or a mixture thereof, by addition of an anti-solvent.
  • the solvent is ethyl acetate.
  • the anti-solvent is water.
  • the anti-solvent is a non-polar organic solvent.
  • the non-polar organic solvent is a hydrocarbon solvent.
  • the anti-solvent is heptane.
  • the solvent is ethyl acetate and the anti-solvent is heptane.
  • the final ratio of solvent to anti-solvent is from about 1:2 to about 1:6.
  • the final ratio of solvent to anti-solvent is from about 1:3 to about 1:5.
  • the final ratio of solvent to anti-solvent is about 1:4.
  • the anti-solvent is added to the solvent at a temperature above room temperature. In one embodiment, the anti-solvent is added at a temperature of from about 30° C. to about 60° C. In one embodiment, the temperature is about 50° C.
  • the non-Form 2 solid form is exposed to one solvent. In one embodiment, the non-Form 2 solid form is exposed to a mixture of two solvents. In one embodiment, the non-Form 2 solid form is exposed to one or more solvents.
  • the solvent is an organic solvent. In one embodiment, the solvent is MTBE, MEK, 1-butanol, 2-methyl-1-propanol, 3-methyl-1-butanol acetone, cyclopentyl methyl ether, or tAmyl alcohol. In one embodiment, the solvent is MEK. In one embodiment, the solvent is MTBE. In one embodiment, the solvent is 1-butanol.
  • the non-Form 2 solid form is an amorphous form of a compound of Formula (I).
  • the period of time sufficient to convert at least about 50% of the total amount of the non-Form 2 solid form into Form 2 is about 1 hr, about 2 hr, about 5 hr, about 10 hr, about 12 hr, about 20 hr, about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 2 of a compound of Formula (I) is prepared by exposing a composition comprising a compound of Formula (I) to one or more solvent as described in the experiments provided herein, including but not limited to evaporation, anti-solvent addition, slow cooling, crash cooling, temperature cycling or solvent drop grinding.
  • Form 2 of a compound of Formula (I) is obtained by evaporation from 1-butanol.
  • Form 2 of a compound of Formula (I) is prepared by temperature cycling a slurry of amorphous Compound 1 in MTBE in an incubator shaker.
  • the temperature is cycled between ambient temperature and an elevated temperature of about 30-60° C.
  • the temperature is cycled at intervals of between about 2 and about 6 hours for a period of about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 2 of a compound of Formula (I) is prepared by temperature cycling a slurry of amorphous Compound 1 in MTBE in an incubator shaker between ambient temperature and a temperature of about 40° C. at intervals of about 4 hours for a period of about 24 hours.
  • the non-Form 3 solid form is exposed to one solvent. In one embodiment, the non-Form 3 solid form is exposed to a mixture of two solvents. In one embodiment, the non-Form 3 solid form is exposed to one or more solvents. In one embodiment, the solvent is water. In one embodiment, the solvent is an organic solvent. In one embodiment, the solvent is methanol or isopropyl acetate. In one embodiment, the mixture of two solvents is a mixture of acetonitrile and heptane. In one embodiment, the non-Form 3 solid form is an amorphous form of a compound of Formula (I).
  • the period of time sufficient to convert at least about 50% of the total amount of the non-Form 3 solid form into Form 3 is about 1 hr, about 2 hr, about 5 hr, about 10 hr, about 12 hr, about 20 hr, about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 3 of a compound of Formula (I) is prepared by exposing a composition comprising a compound of Formula (I) to one or more solvent as described in the experiments provided herein, including but not limited to evaporation, anti-solvent addition, slow cooling, crash cooling, temperature cycling or solvent drop grinding.
  • the non-Form 4 solid form is exposed to one solvent. In one embodiment, the non-Form 4 solid form is exposed to a mixture of two solvents. In one embodiment, the non-Form 4 solid form is exposed to one or more solvents. In one embodiment, the solvent is an organic solvent. In one embodiment, the solvent is acetonitrile. In one embodiment, the non-Form 4 solid form is an amorphous form of a compound of Formula (I).
  • the period of time sufficient to convert at least about 50% of the total amount of the non-Form 4 solid form into Form 4 is about 1 hr, about 2 hr, about 5 hr, about 10 hr, about 12 hr, about 20 hr, about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 4 of a compound of Formula (I) is prepared by exposing a composition comprising a compound of Formula (I) to one or more solvent as described in the experiments provided herein, including but not limited to evaporation, anti-solvent addition, slow cooling, crash cooling, temperature cycling or solvent drop grinding.
  • Form 4 of a compound of Formula (I) is obtained from addition of heptane anti-solvent to a composition comprising amorphous Compound 1 and acetonitrile.
  • Form 4 of a compound of Formula (I) is prepared by temperature cycling a slurry of amorphous Compound 1 in acetonitrile in an incubator shaker. In some embodiments, the temperature is cycled between ambient temperature and an elevated temperature of about 30-60° C.
  • the temperature is cycled at intervals of between about 2 and about 6 hours for a period of about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 4 of a compound of Formula (I) is prepared by temperature cycling a slurry of amorphous Compound 1 in acetonitrile in an incubator shaker between ambient temperature and a temperature of about 40° C. at intervals of about 4 hours for a period of about 72 hours.
  • the non-Form 5 solid form is exposed to one solvent. In one embodiment, the non-Form 5 solid form is exposed to a mixture of two solvents. In one embodiment, the non-Form 5 solid form is exposed to one or more solvents. In one embodiment, the solvent is an organic solvent. In one embodiment, the solvent is acetonitrile. In one embodiment, the non-Form 5 solid form is an amorphous form of a compound of Formula (I).
  • the period of time sufficient to convert at least about 50% of the total amount of the non-Form 5 solid form into Form 5 is about 1 hr, about 2 hr, about 5 hr, about 10 hr, about 12 hr, about 20 hr, about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 5 of a compound of Formula (I) is prepared by exposing a composition comprising a compound of Formula (I) to one or more solvent as described in the experiments provided herein, including but not limited to evaporation, anti-solvent addition, slow cooling, crash cooling, temperature cycling or solvent drop grinding.
  • Form 5 of a compound of Formula (I) is obtained from solvent drop grinding amorphous Compound 1 in acetonitrile.
  • a process for preparing Form 6 of a compound of Formula (I) comprising:
  • the non-Form 6 solid form is exposed to one solvent. In one embodiment, the non-Form 6 solid form is exposed to a mixture of two solvents. In one embodiment, the non-Form 6 solid form is exposed to one or more solvents. In one embodiment, the solvent is an organic solvent. In one embodiment, the solvent is 1,4-dioxane. In one embodiment, the non-Form 6 solid form is an amorphous form of a compound of Formula (I).
  • the period of time sufficient to convert at least about 50% of the total amount of the non-Form 6 solid form into Form 6 is about 1 hr, about 2 hr, about 5 hr, about 10 hr, about 12 hr, about 20 hr, about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 6 of a compound of Formula (I) is prepared by exposing a composition comprising a compound of Formula (I) to one or more solvent as described in the experiments provided herein, including but not limited to evaporation, anti-solvent addition, slow cooling, crash cooling, temperature cycling or solvent drop grinding.
  • Form 6 of a compound of Formula (I) is prepared by temperature cycling a slurry of amorphous Compound 1 in 1,4-dioxane in an incubator shaker.
  • the temperature is cycled between ambient temperature and an elevated temperature of about 30-60° C.
  • the temperature is cycled at intervals of between about 2 and about 6 hours for a period of about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 6 of a compound of Formula (I) is prepared by temperature cycling a slurry of amorphous Compound 1 in 1,4-dioxane in an incubator shaker between ambient temperature and a temperature of about 40° C. at intervals of about 4 hours for a period of about 72 hours.
  • the non-Form 7 solid form is exposed to one solvent. In one embodiment, the non-Form 7 solid form is exposed to a mixture of two solvents. In one embodiment, the non-Form 7 solid form is exposed to one or more solvents.
  • the solvent is an organic solvent. In one embodiment, the solvent is a mixture of 2-butanol and octane, a mixture of methylisobutyl ketone and octane, a mixture of isopropyl acetate and octane, or a mixture of 1,2-dimethoxyethane and octane. In one embodiment, the non-Form 7 solid form is Form 1.
  • the period of time sufficient to convert at least about 50% of the total amount of the non-Form 7 solid form into Form 7 is about 1 hr, about 2 hr, about 5 hr, about 10 hr, about 12 hr, about 20 hr, about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • Form 7 of a compound of Formula (I) is prepared by exposing a composition comprising a compound of Formula (I) to one or more solvent as described in the experiments provided herein, including but not limited to evaporation, anti-solvent addition, slow cooling, crash cooling, temperature cycling or solvent drop grinding.
  • Form 7 of a compound of Formula (I) is prepared by slurrying Form 1 in a solvent at elevated temperature.
  • the solvent is a mixture of 2-butanol and octane, a mixture of methylisobutyl ketone and octane, a mixture of isopropyl acetate and octane, or a mixture of 1,2-dimethoxyethane and octane.
  • the temperature is greater than about 60° C. In one embodiment, the temperature is about 60° C. In one embodiment, the temperature is greater than about 80° C. or higher. In one embodiment, the temperature is about 80° C.
  • Form 7 of a compound of Formula (I) comprises heating a solid form of Form 1 to Form 6, optionally under vacuum.
  • Form 7 of a compound of Formula (I) is prepared by heating Form 2 in a reaction vessel under vacuum.
  • the reaction vessel is a glass vial, wherein Form 2 is spread over one of the vial walls so as to have a large surface area to promote the homogeneous desolvation.
  • the vaccum pressure is between about 2 mbar and about 50 mbar. In some embodiments, the vacuum pressure is about 10 mbar.
  • Form 7 of a compound of Formula (I) is prepared by heating Form 2 under vacuum at a reaction temperature between about 200° C. and about 300° C. In some embodiments, the reaction temperature is about 250° C. In some embodiments, the reaction vessel is maintained at the reaction temperature for a time of between about 3 and about 10 minutes. In some embodiments, the time is about 6 minutes. In some embodiments, the reaction temperature is reached by heating the reaction vessel at a rate of between about 10° C./min and about 30° C./min. In some embodiments, the rate is about 20° C./min. In some embodiments, after the reaction vessel has been heated under vacuum for the desired time, the reaction vessel and/or the solids contained therein are actively cooled. In some embodiments, the active cooling involves use of wet towels.
  • a solid form comprising a salt of a compound of Formula (I):
  • the molar ratio of Compound 1 to a counterion of the salt of Compound 1 may be about 1:1, about 1:2, about 1:3, or about 1:4. In one embodiment, the molar ratio of Compound 1 to counterion is about 1:1. In one embodiment, the molar ratio of Compound 1 to a counterion is about 1:2. In one embodiment, the molar ratio of Compound 1 to a counterion is about 1:3. In one embodiment, the molar ratio of Compound 1 to a counterion is about 1:4.
  • the couterion is chloride, phosphate, besylate, mesylate, citrate or maleate.
  • the salt of Compound 1 is a besylate salt of Compound 1.
  • the salt of Compound 1 is a phosphate salt of Compound 1.
  • a solid form comprising a salt of Compound 1.
  • an unsolvated solid form comprising a salt of Compound 1.
  • an anhydrous solid form comprising a salt of Compound 1.
  • a solvated solid form comprising a salt of Compound 1.
  • a hydrate solid form comprising a salt of Compound 1.
  • a salt of Compound 1 can exist in a variety of solid forms. Such solid forms include crystalline solids or mixtures of crystalline and amorphous solids. In one embodiment, the solid form is substantially crystalline. In one embodiment, the solid form is crystalline.
  • the molar ratio of the salt of Compound 1 to the solvent (e.g. water) in the solid form ranges from about 10:1 to about 1:10. In some embodiments, the molar ratio of the salt of Compound 1 to the solvent (e.g. water) in the solid form ranges from about 5:1 to about 1:5. In some embodiments, the molar ratio of the salt of Compound 1 to the solvent (e.g. water) in the solid form ranges from about 3:1 to about 1:3. In some embodiments, the molar ratio of the sale of Compound 1 to the solvent (e.g. water) in the solid form ranges from about 2:1 to about 1:2. In one embodiment, the molar ratio is about 1:1 (i.e., mono-solvate or mono-hydrate).
  • a Form A of a besylate salt of Compound 1 is provided herein.
  • a representative XRPD pattern of Form A of a besylate salt of Compound 1 is provided in FIG. 24 .
  • a solid form comprising a besylate salt of Compound 1, characterized by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all of the XRPD peaks located at approximately the following positions (e.g., degrees 2 ⁇ 0.2) when measured using Cu K ⁇ radiation: 4.6, 10.7, 11.5, 14.6, 15.0, 17.9, 19.4, 20.4, 21.0, 21.3, 21.4, 23.0, 23.4, and 25.7 2 ⁇ .
  • the solid form is characterized by 3 of the peaks.
  • the solid form is characterized by 5 of the peaks.
  • the solid form is characterized by 7 of the peaks.
  • the solid form is characterized by 9 of the peaks.
  • the solid form is characterized by 11 of the peaks.
  • the solid form is characterized by all of the peaks.
  • a solid form comprising a besylate salt of Compound 1, characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 4.6, 10.7, 11.5, 14.6, 15.0, 17.9, 19.4, 21.0, 21.4, 23.0, and 25.7 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 4.6, 10.7, 11.5, 14.6, 15.0, 17.9, 19.4, 21.0, 21.4, 23.0, and 25.7 2 ⁇ .
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 4.6, 10.7, 11.5, 14.6, 15.0, 17.9, 19.4, 21.0, 21.4, 23.0, and 25.7 2 ⁇ .
  • a solid form comprising a besylate salt of Compound 1, characterized by an XRPD pattern comprising peaks at approximately (e.g., ⁇ 0.2°) 15.0, 17.9 and 23.0° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 10.7 and 14.6° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 4.6 and 25.7° 2 ⁇ .
  • the XRPD pattern comprises peaks at approximately (e.g., ⁇ 0.2°) 4.6, 10.7, 11.5, 14.6, 15.0, 17.9, 21.4, 23.0, and 25.7° 2 ⁇ .
  • a solid form comprising a besylate salt of Compound 1, characterized by an XRPD pattern that matches the XRPD pattern depicted in FIG. 24 .
  • an XRPD pattern described herein is obtained using Cu K ⁇ radiation.
  • the XRPD pattern is measured by XRPD using Cu K ⁇ radiation comprising K ⁇ 1 radiation having a wavelength of 1.5406 ⁇ and K ⁇ 2 radiation having a wavelength of 1.5444 ⁇ , wherein the K ⁇ 1 :K ⁇ 2 ratio is 0.5.
  • FIG. 25 A representative overlay of TGA/DSC thermograms of Form A of a besylate salt of Compound 1 is provided in FIG. 25 .
  • a solid form comprising a besylate salt of Compound 1, which exhibits a weight loss of about 2.8% upon heating from about 25° C. to about 125° C., and/or a weight loss of about 1.2% upon heating from about 125° C. to about 275° C.
  • a solid form comprising a besylate salt of Compound 1, which exhibits a decomposition upon heating from about 25° C. (e.g. ⁇ 2°) and/or a decomposition upon heating from about 125° C. (e.g. ⁇ 2°).
  • the solid form is characterized by a TGA thermogram that matches the TGA thermogram depicted in FIG. 25 .
  • the TGA thermogram is as measured using a heating rate of about 10° C./minute.
  • a representative DSC thermograms of Form A of a besylate salt of Compound 1 is provided in FIG. 26 .
  • a solid form comprising a besylate salt of Compound 1, which exhibits, as characterized by DSC, a thermal event (endo) with an onset temperature at about 128° C. (e.g. ⁇ 2°).
  • the thermal event also has a peak temperature of about 136° C. (e.g. ⁇ 2°).
  • the thermal event corresponds to melting.
  • the solid form is characterized by a DSC thermogram that matches the DSC thermogram depicted in FIG. 26 .
  • the DSC thermogram is as measured by DSC using a scanning rate of about 10° C./minute.
  • a representative DVS isotherm of Form A of a besylate salt of Compound 1 is provided in FIG. 27 .
  • a solid form comprising a besylate salt of Compound 1 which exhibits a weight increase of about 2% (e.g. ⁇ 0.05%) when subjected to an increase in relative humidity from about 0 to about 90% relative humidity.
  • the solid form is characterized by a DVS isotherm that matches the DVS isotherm depicted in FIG. 27 .
  • the DVS isotherm is as measured at about 25° C.
  • a solid form comprising a besylate salt of Compound 1.
  • the solid form is a monobesylate salt of Compound 1.
  • the solid form is a hydrate of a besylate salt of Compound 1.
  • the solid form is a monohydrate of a besylate salt of Compound 1.
  • the solid form is a crystalline solid form comprising a besylate salt of Compound 1.
  • the solid form is substantially free of amorphous Compound 1 and/or an amorphous form of a salt of Compound 1.
  • the solid form comprising a besylate salt of Compound 1 is substantially free of other solid forms (i.e., crystalline forms) of Compound 1.
  • the solid form is provided as substantially pure.
  • the solid form is substantially chemically pure.
  • the solid form is substantially chemically pure.
  • provided herein is a solid form comprising Form A of a besylate salt of Compound 1 and amorphous free base of Compound 1. In one embodiment, provided herein is a solid form comprising Form A of a besylate salt of Compound 1 and amorphous besylate salt of Compound 1. In one embodiment, provided herein is a solid form comprising Form A of a besylate salt of Compound 1 and one or more other crystalline forms of a besylate salt of Compound 1.
  • a Form A of a phosphate salt of Compound 1 is provided herein.
  • a representative XRPD pattern of Form A of a phosphate salt of Compound 1 is provided in FIG. 28 .
  • a solid form comprising a phosphate salt of Compound 1, characterized by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or all of the XRPD peaks located at approximately the following positions (e.g., degrees 2 ⁇ 0.2) when measured using Cu K ⁇ radiation: 10.8, 14.2, 15.0, 15.9, 17.7, 18.5, 19.4, 20.1, 20.7, 21.6, 22.3, 24.1, 24.8, and 25.8 2 ⁇ .
  • the solid form is characterized by 3 of the peaks.
  • the solid form is characterized by 5 of the peaks.
  • the solid form is characterized by 7 of the peaks.
  • the solid form is characterized by 9 of the peaks.
  • the solid form is characterized by 11 of the peaks.
  • the solid form is characterized by all of the peaks.
  • a solid form comprising a phosphate salt of Compound 1, characterized by an XRPD pattern, when measured using Cu K ⁇ radiation, comprising at least three peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 10.8, 14.2, 15.0, 15.9, 17.7, 18.5, 19.4, 22.3, 24.1, 24.8, and 25.8.
  • the solid form is characterized by an XRPD pattern comprising at least four peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 10.8, 14.2, 15.0, 15.9, 17.7, 18.5, 19.4, 22.3, 24.1, 24.8, and 25.8.
  • the solid form is characterized by an XRPD pattern comprising at least five peaks selected from the group consisting of approximately (e.g., ⁇ 0.2°) 10.8, 14.2, 15.0, 15.9, 17.7, 18.5, 19.4, 22.3, 24.1, 24.8, and 25.8.
  • a solid form comprising a phosphate salt of Compound 1, characterized by an XRPD pattern comprising peaks at approximately (e.g., ⁇ 0.2°) 10.8, 18.5, and 24.8° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 22.3 and 24.1° 2 ⁇ .
  • the solid form is characterized by an XRPD pattern further comprising peaks at approximately (e.g., ⁇ 0.2°) 14.2 and 17.7° 2 ⁇ .
  • the XRPD pattern comprises peaks at approximately (e.g., ⁇ 0.2°) 10.8, 14.2, 15.9, 17.7, 18.5, 22.3, 24.1, and 24.8° 2 ⁇ .
  • a solid form comprising a phosphate salt of Compound 1, characterized by an XRPD pattern that matches the XRPD pattern depicted in FIG. 28 .
  • an XRPD pattern described herein is obtained using Cu K ⁇ radiation.
  • the XRPD pattern is measured by XRPD using Cu K ⁇ radiation comprising K ⁇ 1 radiation having a wavelength of 1.5406 ⁇ and K ⁇ 2 radiation having a wavelength of 1.5444 ⁇ , wherein the K ⁇ 1 :K ⁇ 2 ratio is 0.5.
  • FIG. 29 A representative overlay of TGA/DSC thermograms of Form A of a phosphate salt of Compound 1 is provided in FIG. 29 .
  • a solid form comprising a phosphate salt of Compound 1, which exhibits a weight loss of about 5.8% upon heating from about 25° C. to about 175° C.
  • a solid form comprising a phosphate salt of Compound 1, which exhibits decomposition upon heating from about 25° C. (e.g. ⁇ 2°).
  • the solid form comprising a phosphate salt of Compound 1 is characterized by a TGA thermogram that matches the TGA thermogram depicted in FIG. 29 .
  • the TGA thermogram is as measured using a heating rate of about 10° C./minute.
  • solid form comprising a phosphate salt of Compound 1.
  • the solid form comprises a single phosphate counterion.
  • the solid form is a hydrate of a phosphate salt of Compound 1.
  • the solid form is a solvate of a phosphate salt of Compound 1.
  • the solid form is a crystalline solid form comprising a phosphate salt of Compound 1.
  • the solid form is substantially free of amorphous Compound 1 and/or an amorphous form of a salt of Compound 1.
  • the solid form comprising a phosphate salt of Compound 1 is substantially free of other solid forms (e.g., crystalline forms) of Compound 1.
  • the solid form is provided as substantially pure.
  • the solid form is substantially chemically pure.
  • the solid form is substantially physically pure.
  • a solid form comprising Form A of a phosphate salt of Compound 1 and amorphous free base of Compound 1. In one embodiment, provided herein is a solid form comprising Form A of a phosphate salt of Compound 1 and amorphous phosphate salt of Compound 1. In one embodiment, provided herein is a solid form comprising Form A of a phosphate salt of Compound 1 and one or more other crystalline forms of a phosphate salt of Compound 1.
  • Form 1 of a free base of Compound 1 is exposed to benezenesulfonic acid in one or more solvents.
  • amorphous Compound 1 is exposed to benezenesulfonic acid in one or more solvents.
  • mostly amorphous Compound 1 is exposed to benezenesulfonic acid in one or more solvents.
  • Compound 1 is exposed an aqueous benzenesulfonic acid solution.
  • the benesulfonic acid solution has a concentration of between about 0.1M and 5M benezenesulfonic acid.
  • the benesulfonic acid solution has a concentration of 1M benezenesulfonic acid.
  • Compound 1 is exposed about 0.8 to about 1.3 molar equivalents of benzenesulfonic acid. In one embodiment, Compound 1 is exposed about 1.1 molar equivalents of benzenesulfonic acid. In one embodiment, benezenesulfonic acid is added to Compound 1 in a one or more solvents. In one embodiment, Compound 1 is dissolved in a solvent. In one embodiment, Compound 1 is dissolved in a solvent at a concentration of between about 10 and about 200 mg/ml of Compound 1. In one embodiment, Compound 1 is dissolved in a solvent at a concentration of about 100 mg/ml of Compound 1. In one embodiment, the solvent is an organic solvent. In one embodiment, the solvent is an alcohol.
  • the solvent is 1-butanol, 1-propanol, 2-methyl-1-propanol, 2-butanol, ethanol, or tAmyl alcohol.
  • the solvent is tetrahydrofuran, 2-methyltetrahydrofuran (2-MeTHF), butyl acetate, cyclopentyl methyl ether (CPME), ethyl acetate, isopropyl acetate, methyl ethyl ketone, toluene, methylisobutyl ketone or methyl acetate.
  • the solvent is 2-MeTHF.
  • the composition comprising Compound 1 is agitated following exposure to benezenesulfonic acid in one or more solvents.
  • the period of time sufficient to convert at least about 50% of the total amount of Compound 1 into a solid form of a besylate salt of Compound 1 is less than 1 hr, about 1 hr, about 2 hr, about 5 hr, about 10 hr, about 12 hr, about 20 hr, about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • the process of preparing a solid form of a besylate salt of Compound 1 further comprises a step of crystallizing or recrystallizing from a solvent after exposing a composition comprising Compound 1 to benezenesulfonic acid.
  • the crystallization or recrystallization step includes but not limited to evaporation, anti-solvent addition, slow cooling, crash cooling, temperature cycling or solvent drop grinding.
  • the process of preparing the solid form of a besylate salt of Compound 1 further comprises a step of evaporating solvent after exposing a composition comprising Compound 1 to benezenesulfonic acid.
  • the process comprises a step of evaporating 2-MeTHF solvent.
  • Form 1 of a free base of Compound 1 is exposed to phosphoric acid in one or more solvents.
  • amorphous Compound 1 is exposed to phosphoric acid in one or more solvents.
  • mostly amorphous Compound 1 is exposed to phosphoric acid in one or more solvents.
  • Compound 1 is exposed an aqueous phosphoric acid solution.
  • the phosphoric acid solution has a concentration of between about 0.1M and 15M phosphoric acid.
  • the phosphoric acid solution has a concentration of 1M phosphoric acid.
  • Compound 1 is exposed about 0.8 to about 1.3 molar equivalents of phosphoric acid.
  • Compound 1 is exposed about 1.1 molar equivalents of phosphoric acid.
  • phosphoric acid is added to Compound 1 in a one or more solvents.
  • Compound 1 is dissolved in a solvent.
  • Compound 1 is dissolved in a solvent at a concentration of between about 10 and about 200 mg/ml of Compound 1.
  • Compound 1 is dissolved in a solvent at a concentration of about 100 mg/ml of Compound 1.
  • the solvent is an organic solvent.
  • the solvent is an alcohol.
  • the solvent is 1-butanol, 1-propanol, 2-methyl-1-propanol, 2-butanol, ethanol, or tAmyl alcohol.
  • the solvent is tetrahydrofuran, 2-methyltetrahydrofuran (2-MeTHF), butyl acetate, cyclopentyl methyl ether (CPME), ethyl acetate, isopropyl acetate, methyl ethyl ketone, toluene, methylisobutyl ketone or methyl acetate.
  • the solvent is 2-MeTHF.
  • the composition comprising Compound 1 is agitated following exposure to phosphoric acid in one or more solvents.
  • the period of time sufficient to convert at least about 50% of the total amount of Compound 1 into a solid form of a phosphate salt of Compound 1 is less than 1 hr, about 1 hr, about 2 hr, about 5 hr, about 10 hr, about 12 hr, about 20 hr, about 24 hr, about 30 hr, about 40 hr, about 48 hr, about 72 hr, about 97 hours, about 121 hours, or greater than 121 hours.
  • the process of preparing a solid form of a phosphate salt of Compound 1 further comprises a step of crystallizing or recrystallizing from a solvent after exposing a composition comprising Compound 1 to phosphoric acid.
  • the crystallization or recrystallization step includes but not limited to evaporation, anti-solvent addition, slow cooling, crash cooling, temperature cycling or solvent drop grinding.
  • the process of preparing the solid form of a phosphate salt of Compound 1 further comprises a step of evaporating solvent after exposing a composition comprising Compound 1 to phosphoric acid.
  • the process comprises a step of evaporating 2-MeTHF solvent.
  • step 1.0 cyclizing a compound of Formula (II):
  • step 1.0 occurs in the presence of a base.
  • the base is an organic base.
  • the organic base is a carboxylate base.
  • the carboxylate base is lithium acetate, sodium acetate, potassium acetate, lithium pivalate, sodium pivalate, potassium pivalate, cesium acetate, or cesium pivalate.
  • the base is potassium acetate.
  • the base is potassium pivalate.
  • the molar ratio of the compound of Formula (II) to base in step 1.0 is from about 1:2 to about 1:6. In one embodiment, the molar ratio of the compound of Formula (II) to base in step 1.0 is about 1:5. In one embodiment, the molar ratio of the compound of Formula (II) to base in step 1.0 is about 1:3.
  • step 1.0 occurs in the presence of a catalyst precursor.
  • the catalyst precursor comprises a palladium source.
  • the palladium source is Pd-G3, Pd 2 (dba) 3 , PdCl 2 (MeCN) 2 , Pd(OAc) 2 , Pd(PPh 3 ) 4 , PdCl 2 (PPh 3 ) 2 , PdCl 2 (PCy 3 ) 2 , PdCl 2 (dtbpf), PdCl 2 (dppf), PdCl 2 (Amphos), ⁇ Pd( ⁇ -Br)[P(tBu) 3 ] ⁇ 2 , PdCl 2 [P(Cy) 3 ] 2 , Pd[P(tBu) 3 ] 2 , PdCl 2 (dtbpf), Pd[P(Cy) 3 ] 2 , or PdCl 2 [P(tBu)(Cy) 2 ] 2 , or PdCl 2 [P(
  • the palladium source is Pd(OAc) 2 .
  • the catalyst precursor comprises Pd(OAc) 2 .
  • the catalysts precursor comprises a ligand.
  • the ligand is a phosphine ligand or bisphosphine ligand.
  • the ligand is phosphine or bisphosphine ligand commonly used in the art.
  • the ligand is a cataCXium ligand.
  • the cataCXium ligand is cataCXium A, cataCXium ABn, cataCXium AHI, cataCXium PIntB, cataCXium PICy, cataCXium PtB, cataCXium POmeB, or cataCXium C.
  • the cataCXium ligand is cataCXium A.
  • the catalyst precursor comprises cataCXium A.
  • use of a cataCXium A results in improved conversion in step 1.0.
  • the catalyst precursor comprises a palladium source and a ligand.
  • the catalyst precursor and the ligand are pre-formed palladium ligand complexes such as cataCXium A Pd G2, cataCXium A Pd G3, or bis(butyldi-1-adamantylphosphine) palladium diacetate.
  • the catalyst precursor comprises Pd(OAc) 2 and cataCXium A.
  • step 1.0 proceeds to greater than 90%, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or greater than 99% conversion within about 22-24 hours, as determined by HPLC.
  • the molar ratio of the compound of Formula (II) to palladium source in step 1.0 is from about 1:0.01 (i.e. 1 mol %) to about 1:0.15 (i.e. 15 mol %). In some embodiments, the molar ratio of the compound of Formula (II) to palladium source in step 1.0 is about 1:0.02, about 1:0.03, about 1:0.04, about 1:0.05, about 1:0.06, about 1:0.07, about 1:0.08, about 1:0.09, about 1:0.10, about 1:0.11, or about 1:0.12. In one embodiment, the molar ratio of the compound of Formula (II) to palladium source in step 1.0 is about 1:0.02 (i.e.
  • the molar ratio of the compound of Formula (II) to palladium source in step 1.0 is about 1:0.03 (i.e. 3 mol %). In one embodiment, the molar ratio of the compound of Formula (II) to palladium source in step 1.0 is about 1:0.04 (i.e. 4 mol %). In one embodiment, a palladium loading of less than about 10 mol %, less than about 5 mol %, or about 4 mol %, is employed in step 1.0.
  • the molar ratio of the compound of Formula (II) to ligand in step 1.0 is from about 1:0.01 (i.e. 1 mol %) to about 1:0.15 (i.e. 15 mol %). In some embodiments, the molar ratio of the compound of Formula (II) to ligand in step 1.0 is about 1:0.03, about 1:0.04, about 1:0.05, about 1:0.06, about 1:0.07, about 1:0.08, about 1:0.09, about 1:0.10, about 1:0.11, or about 1:0.12. In one embodiment, the molar ratio of the compound of Formula (II) to ligand in step 1.0 is about 1:0.04 (i.e. 4 mol %).
  • the molar ratio of the compound of Formula (II) to ligand in step 1.0 is about 1:0.08 (i.e. 8 mol %). In one embodiment, a ligand loading of less than about 10 mol % is employed in step 1.0.
  • the molar ratio of the ligand to the palladium source is from about 5:1 about 1:5. In one embodiment, the molar ratio of the ligand to the palladium source is from about 2:1 to about 1:2. In one embodiment, the molar ratio of the ligand to the palladium source is from about 2:1 to about 1:1. In one embodiment, the ligand is a monodentate ligand and the molar ratio of the ligand to the palladium source is about 2:1. In one embodiment, the ligand is a monodentate ligand and the molar ratio of the ligand to the palladium source is about 1:1.
  • the ligand is a bidentate ligand and the molar ratio of the ligand to the palladium source is about 1:1. In one embodiment, the ligand is a bidentate ligand and the molar ratio of the ligand to the palladium source is about 1:2.
  • Step 1.0 may occur in a solvent suitable for the reaction.
  • the solvent is an organic solvent or a mixture of organic solvents.
  • the solvent is a high-boiling solvent, including but not limited to C 4-12 aliphatic alcohol (branched or unbranched), toluene, anisole, 2-MeTHF, DMF, NMP, DMA or tAmOH.
  • the solvent is an alcohol.
  • the solvent is t-amyl alcohol (tAmOH).
  • the solvent is n-BuOH, s-BuOH, or t-BuOH.
  • the volume of solvent in step 1.0 is from about 10 vol to about 30 vol. In one embodiment, the volume of the solvent in step 1.0 is about 20 vol. In some embodiments, the volume of solvent in step 1.0 is from about 5 vol to about 10 vol. In one embodiment, the volume of solvent in step 1.0 is about 8 vol.
  • vol refers to the volume (L or mL) of a solvent relevant to the weight (kg or g respectivley) of the limiting reagent.
  • step 1.0 occurs in an inert atmosphere (i.e. under conditions which eliminate or substantially reduce the presence of atmospheric oxygen).
  • the solvent is sparged with an inert gas (e.g. dinitrogen or argon) in step 1.0.
  • step 1.0 occurs at a reaction temperature of from about 90° C. to about 120° C.
  • the reaction temperature is the boiling temperature of the solvent.
  • the reaction temperature is from about 100° C. to about 110° C.
  • the reaction temperature is about 102° C.
  • the reaction temperature is about 105° C.
  • step 1.0 occurs at a reaction time from about 16 hours to about 30 hours. In one embodiment, the reaction time is from about 22 hours to about 24 hours.
  • step 1.0 occurs in the presence of potassium pivalate base and a catalyst precursor comprising Pd(OAc) 2 and cataCXium A.
  • the molar ratios of the compound of Formula (I) to potassium pivalate, Pd(OAc) 2 and cataCXium A are 1:3, 1:0.04, and 1:0.08, respectively.
  • step 1.0 occurs in a solvent of t-amyl alcohol and a solvent volume of 20 vol at a reaction temperature of about 102° C.
  • the molar ratios of the compound of Formula (I) to potassium pivalate, Pd(OAc) 2 and cataCXium A are 1:3, 1:0.02, and 1:0.04, respectively.
  • step 1.0 occurs in a solvent of t-amyl alcohol and a solvent volume of 8 vol at a reaction temperature of about 105° C.
  • the solvent is sparged with dinitrogen gas in step 1.0.
  • step 1.0 proceeds to greater than 90%, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or greater than 99% conversion within about 22-24 hours, as determined by HPLC and/or NMR. In some embodiments, step 1.0 provides less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% of an impurity distinct from the compound of Formula (I).
  • Impurities provided in step 1.0 may include, but are not limited to, starting material, a debrominated species of Formula (SP-1), a dimer of Formula (SP-2), a t-butyl carbonylated species of Formula (SP-3), a regioisomer of Formula (SP-7), and/or the compound of Formula (V) described herein below.
  • SP-1 debrominated species of Formula
  • SP-2 dimer of Formula
  • SP-3 t-butyl carbonylated species of Formula
  • SP-7 a regioisomer of Formula
  • the total amount of impurities provided in step 1.0 is less than less than about 10 wt %, less than about 5 wt %, less than about 4 wt %, less than about 3 wt %, less than about 2 wt %, less than about 1 wt %, less than about 0.5 wt %, less than about 0.1 wt %, or less than about 0.05 wt %.
  • step 1.0 further comprises purification of the compound of Formula (I).
  • the compound of Formula (I) produced in step 1.0 is purified by palladium remediation, precipitation from a solvent by an anti-solvent, and/or (re)crystallization.
  • the palladium remediation comprises treatment with any palladium scavenger known in the art.
  • the palladium scavenger is a thiopropyl silica scavenger.
  • the palladium remediation occurs at a temperature above room temperature, e.g., from about 40° C. to about 80° C., e.g., about 60° C.
  • the palladium remediation comprises treatment with a palladium scavenger for a period of greater than 1 hour, greater than 4 hours, greater than 10 hours, greater than 14 hours, greater than 16 hours, or about 18 hours.
  • the palladium remediation comprises more than one treatment with a palladium scavenger, e.g., two treatments with a palladium scavenger.
  • the palladium remediation comprises purifying the compound of Formula (I) by functionalized silica treatment.
  • a thiol functionalized silica gel is used to scavenge palladium from the reaction mixture. In some embodiments, this is done at room temperature. In other embodiments, it is performed at elevated temperatures (60-100° C.).
  • the scavenger loading is up to 60% by wt relative to the product mass. In other embodiments it is 0-30% by wt relative to the product mass.
  • the palladium remediation comprises purifying the compound of Formula (I) by silica gel chromatography.
  • the palladium remediation comprises washing an organic solution of the compound of Formula (I) with aqueous L-cysteine.
  • the aqueous L-cysteine is 5 w % aqueous solution of L-cysteine.
  • the washing is conducted at elevated temperatures (i.e. at a temperature above room temperature), for example at a temperature of about 35 to about 50° C., or about 40 to about 45° C.
  • the palladium remediation comprises purifying the compound of Formula (I) by silica gel chromatography and washing an organic solution of the compound of Formula (I) with aqueous L-cysteine.
  • the compound of Formula (I) is precipitated from a high-boiling organic solvent. In one embodiment, the compound of Formula (I) is precipitated from anisole. In one embodiment, the compound of Formula (I) is precipitated from a solvent by addition of a non-polar organic anti-solvent. In one embodiment, the non-polar organic anti-solvent is a hydrocarbon solvent. In one embodiment, the anti-solvent is heptane. In one embodiment, the final ratio of solvent to anti-solvent is from about 1:2 to about 1:6. In one embodiment, the final ratio of solvent to anti-solvent is from about 1:4 to about 1:6. In one embodiment, the final ratio of solvent to anti-solvent is about 1:5. In one embodiment, the final ratio of solvent to anti-solvent is about 1:5.5.
  • the compound of Formula (I) is crystallized or recrystallized from an organic solvent or a mixture of organic solvents. In one embodiment, crystallization or recrystallization of the compound of Formula (I) provides Form 1 of Compound 1. In one embodiment, the compound of Formula (I) is crystallized or recrystallized from 2-MeTHF, isopropyl acetate, acetone, anisole, ethanol, ethyl acetate, isopropyl acetate, methyl ethyl ketone, or a mixture thereof, optionally by addition of an anti-solvent. In one embodiment, the anti-solvent is water. In one embodiment, the anti-solvent is a non-polar organic solvent.
  • the non-polar organic solvent is a hydrocarbon solvent.
  • the anti-solvent is heptane.
  • the solvent is ethyl acetate and the anti-solvent is heptane.
  • the final ratio of solvent to anti-solvent is from about 1:2 to about 1:6.
  • the final ratio of solvent to anti-solvent is from about 1:3 to about 1:5.
  • the final ratio of solvent to anti-solvent is about 1:4.
  • the anti-solvent is added to the solvent at a temperature above room temperature. In one embodiment, the anti-solvent is added at a temperature of from about 30° C. to about 60° C. In one embodiment, the temperature is about 50° C.
  • step 1.0 provides a compound of Formula (I) in a substantially pure form. In certain embodiments, step 1.0 provides a compound of Formula (I) in a substantially chemically pure form. In certain embodiments, step 1.0 provides a compound of Formula (I) having a chemical and/or physical purity of at least about 97% w/w. In certain embodiments, step 1.0 provides a compound of Formula (I) having a chemical and/or physical purity of at least about 98% w/w. In certain embodiments, step 1.0 provides a compound of Formula (I) having a chemical and/or physical purity of at least about 99% w/w.
  • step 1.0 provides a compound of Formula (I) having a chemical and/or physical purity of about 99.71% w/w. In certain embodiments, step 1.0 provides a compound of Formula (I) having a chemical and/or physical purity of about 99.9% w/w. In certain embodiments, step 1.0 provides a compound of Formula (I) having an enantiomeric purity of at least about 98% (e.g. 99% or 99.5%). In certain embodiments, step 1.0 provides a compound of Formula (I) having an enantiomeric purity of at least about 99.5%. In certain embodiments, step 1.0 provides a compound of Formula (I) having an enantiomeric purity of about 99.9%.
  • step 1.0 provides a compound of Formula (I) having an enantiomeric purity of about 100%. In certain embodiments, step 1.0 provides a compound of Formula (I) substantially free of impurities. In certain embodiments, step 1.0 provides a composition comprising Compound 1 having a residual palladium content of less than about 200 ppm, less than about 100 ppm, less than about 50 ppm, less than about 40 ppm, less than about 30 ppm, less than about 20 ppm, or less than about 10 ppm. In certain embodiments, step 1.0 provides a compound of Formula (I) in a substantially enantiomerically pure form. In certain embodiments, step 1.0 provides a compound of Formula (I) in a substantially physically pure form. In certain embodiments, step 1.0 provides a compound of Formula (I) in a solid form having a desired morphology (e.g. a specific crystalline form, such as Form 1 of Compound 1) or advantageous rheological properties.
  • a desired morphology e.
  • a process of preparing a compound of Formula (II), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof comprising:
  • step 2.0 reacting a compound of Formula (III):
  • the molar ratio of the compound of Formula (III) to the compound of Formula (IV) in step 2.0 is from about 1:1 to about 1:1.5. In one embodiment, the molar ratio of the compound of Formula (III) to the compound of Formula (IV) in step 2.0 is about 1.2. In one embodiment, the molar ratio of the compound of Formula (III) to the compound of Formula (IV) in step 2.0 is about 1.1. In one embodiment, the molar ratio of the compound of Formula (III) to the compound of Formula (IV) in step 2.0 is about 1.14.
  • step 2.0 occurs in the presence of a base. In some embodiments, step 2.0 occurs in the presence of an alkali metal base.
  • the base is an alkali metal hydroxide, carbonate, hydrogencarbonate, phosphate, hydrogenphosphate, or dihydrogenphosphate.
  • the base is LiOH, Li 2 CO 3 , NaOH, KOH, tBuOK, Na 2 CO 3 , K 2 CO 3 , Cs 2 CO 3 , NaHCO 3 , KHCO 3 , Na 3 PO 4 , K 3 PO 4 , Na 2 HPO 4 , K 2 HPO 4 , NaH 2 PO 4 , or KH 2 PO 4 .
  • the base is sodium carbonate (Na 2 CO 3 ).
  • the base is cesium carbonate (Cs 2 CO 3 ). In one embodiment, the base is potassium phosphate (K 3 PO 4 ). In some embodiments, step 2.0 occurs in the presence of a nitrogen containing base. In some embodiments, step 2.0 occurs in the presence of 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU), 1,1,3,3-tetramethylguanidine (TMG), triazabicyclodecene (TBD), or potassium bis(trimethylsilyl)amide (KHMDS).
  • DBU 1,8-diazabicyclo[5.4.0]undec-7-ene
  • TMG 1,1,3,3-tetramethylguanidine
  • TBD triazabicyclodecene
  • KHMDS potassium bis(trimethylsilyl)amide
  • the molar ratio of the compound of Formula (III) to base in step 2.0 is from about 1:1 to about 1:5. In some embodiments, the molar ratio of the compound of Formula (III) to base in step 2.0 is from about 1:1 to about 1:3. In one embodiment, the molar ratio of the compound of Formula (III) to base in step 2.0 is about 1:1. In one embodiment, the molar ratio of the compound of Formula (III) to base in step 2.0 is about 1:2. In one embodiment, the molar ratio of the compound of Formula (III) to base in step 2.0 is about 1:2.2. In one embodiment, the molar ratio of the compound of Formula (III) to base in step 2.0 is about 1:4.1.
  • Step 2.0 may occur in a solvent suitable for the reaction.
  • the solvent is an organic solvent or a mixture of organic solvents.
  • the solvent is a polar organic solvent.
  • the solvent is DMF, DCM, IPAc, ehyl acetate, t-amyl alcohol, 1,4-dioxane, DMA, NMP, ACN, or DMSO.
  • step 2.0 occurs in a solvent of N-methyl-2-pyrrolidone (NMP).
  • NMP N-methyl-2-pyrrolidone
  • step 2.0 occurs in a solvent of dimethyl acetamide (DMA).
  • step 2.0 occurs in a solvent of dimethyl formamide (DMF).
  • the volume of solvent in step 2.0 is from about 4 vol to about 20 vol. In one embodiment, the volume of the solvent in step 2.0 is about 16 vol. In some embodiments, the volume of solvent in step 2.0 is from about 4 vol to about 15 vol. In one embodiment, the volume of the solvent in step 2.0 is 15 vol. In one embodiment, the volume of the solvent in step 2.0 is about 13 vol. In one embodiment, the volume of the solvent in step 2.0 is about 8 vol. In one embodiment, the volume of the solvent in step 2.0 is about 6 vol. In one embodiment, the volume of the solvent in step 2.0 is about 5.6 vol.
  • step 2.0 occurs in an inert atmosphere (i.e. under conditions which eliminate or substantially reduce the presence of atmospheric oxygen).
  • the solvent is sparged with an inert gas (e.g. dinitrogen or argon) in step 2.0.
  • step 2.0 occurs at a reaction temperature of from about 20° C. to about 60° C. In some embodiments, step 2.0 occurs at a reaction temperature of from about 35° C. to about 45° C. In some embodiments, step 2.0 occurs at a reaction temperature of from about 20° C. to about 30° C. In one embodiment, the reaction temperature is about 20° C. In one embodiment, the reaction temperature is about 25° C. In one embodiment, step 2.0 is conducted at room temperature. In one embodiment, step 2.0 is conducted at a temperature that minimizes decomposition of the compound of Formula (III).
  • step 2.0 occurs at a reaction time from about 10 hours to about 30 hours. In one embodiment, the reaction time is about 22 hours.
  • the molar ratio of the compound of Formula (III) to the compound of Formula (IV) in step 2.0 is about 1:1.
  • step 2.0 occurs in the presence of cesium carbonate (Cs 2 CO 3 ) and the molar ratio of the compound of Formula (III) to Cs 2 CO 3 in step 2.0 is about 1:2.2.
  • step 2.0 occurs in a solvent of DMA.
  • step 2.0 occurs in a solvent of NMP.
  • step 2.0 occurs in a solvent of N-methyl-2-pyrrolidone (NMP) and a solvent volume of 6 vol. at room temperature.
  • the solvent is sparged with nitrogen gas in step 2.0.
  • the molar ratio of the compound of Formula (III) to the compound of Formula (IV) in step 2.0 is about 1:1.
  • step 2.0 occurs in the presence of potassium phosphate (K 3 PO 4 ) and the molar ratio of the compound of Formula (III) to K 3 PO 4 in step 2.0 is about 1:4.1.
  • step 2.0 occurs in a solvent of dimethyl formamide (DMA) and a solvent volume of 16 vol. at about 35° C. to about 45° C.
  • the solvent is sparged with dinitrogen gas in step 2.0.
  • the molar ratio of the compound of Formula (III) to the compound of Formula (IV) in step 2.0 is about 1:14.
  • step 2.0 occurs in the presence of cesium carbonate (Cs 2 CO 3 ) and the molar ratio of the compound of Formula (III) to Cs 2 CO 3 in step 2.0 is about 1:2.2.
  • step 2.0 occurs in a solvent of N-methyl-2-pyrrolidone (NMP) and a solvent volume of 5.6 vol. at about 20° C. to about 30° C.
  • NMP N-methyl-2-pyrrolidone
  • the solvent is sparged with dinitrogen gas in step 2.0.
  • step 2.0 proceeds to greater than 90%, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or greater than 99% conversion within about 22-24 hours, as determined by HPLC and/or NMR. In some embodiments, step 2.0 provides less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% of an impurity distinct from the compound of Formula (II). Impurities provided in step 2.0 may include, but are not limited to, starting material, an elimination product of Formula (SP-5) and/or the compound of Formula (V) described herein below.
  • the total amount of impurities provided in step 2.0 is less than less than about 10%, less than about 5 wt %, less than about 4 wt %, less than about 3 wt %, less than about 2 wt %, or less than about 1 wt %.
  • step 2.0 further comprises purification of the compound of Formula (II).
  • the compound of Formula (II) produced in step 2.0 is purified by precipitation from a solvent by an anti-solvent and/or (re)crystallization.
  • the compound of Formula (II) is precipitated from an organic solvent. In one embodiment, the compound of Formula (II) is precipitated from NMP. In one embodiment, the compound of Formula (II) is precipitated from a solvent by addition of an anti-solvent. In one embodiment, the anti-solvent is water. In one embodiment, the final volume ratio of solvent to anti-solvent is from about 1:1 to about 1:4. In one embodiment, the final volume ratio of solvent to anti-solvent is about 1:1.5. In one embodiment, the final volume ratio of solvent to anti-solvent is about 1:2. In one embodiment, the final volume ratio of solvent to anti-solvent is about 1:3.
  • the compound of Formula (II) is crystallized or recrystallized from an organic solvent or a mixture of organic solvents.
  • the compound of Formula (II) is crystallized or recrystallized from 2-MeTHF, isopropyl acetate, acetone, anisole, toluene, t-butanol, acetonitrile, ethanol, ethyl acetate, isopropyl acetate, methyl ethyl ketone, or a mixture thereof, optionally by addition of an anti-solvent.
  • the anti-solvent is water.
  • the anti-solvent is a non-polar organic solvent.
  • the non-polar organic solvent is a hydrocarbon solvent.
  • the anti-solvent is heptane. In one embodiment, the anti-solvent is methylcyclohexane. In one embodiment, the solvent is ethyl acetate, toluene, anisole, t-butanol, acetonitrile, isopropyl acetate, or EtOH, and the anti-solvent is water, heptane or methylcyclohexane. In one embodiment, the solvent is ethyl acetate and the anti-solvent is heptane. In one embodiment, the solvent is toluene and the anti-solvent is heptane.
  • the solvent is a mixture of ethyl acetate and toluene, and the anti-solvent is heptane.
  • the final volume ratio of solvent to anti-solvent is from about 1:2 to about 1:6.
  • the final volume ratio of solvent to anti-solvent is from about 1:3 to about 1:5.
  • the final volume ratio of solvent to anti-solvent is about 1:3.
  • the final volume ratio of solvent to anti-solvent is about 1:4.
  • the final volume ratio of solvent to anti-solvent is about 1:5.
  • the anti-solvent is added to the solvent at a temperature above room temperature. In one embodiment, the anti-solvent is added at a temperature of from about 30° C. to about 60° C. In one embodiment, the temperature is about 40° C.
  • step 2.0 provides a compound of Formula (II) in a substantially pure form. In certain embodiments, step 2.0 provides a compound of Formula (II) in a substantially chemically pure form. In certain embodiments, step 2.0 provides a compound of Formula (II) in a substantially enantiomerically pure form. In certain embodiments, step 2.0 provides a compound of Formula (II) substantially free of impurities and easy scale up. In certain embodiments, step 2.0 reduces, eliminates or minimizes the amount of impurities carried forward into step 1.0.
  • a process for preparing a compound of Formula (III), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof comprising:
  • step 3.0 reacting a compound of Formula (V):
  • the mesylating reagent in step 3.0 is mesyl chloride (MsCl) or methanesulfonic anhydride (Ms 2 O). In one embodiment, the mesylating reagent is methanesulfonic anhydride (Ms 2 O).
  • the molar ratio of the compound of Formula (V) to mesylating reagent in step 3.0 is from about 1:1 to about 1:2. In one embodiment, the molar ratio of the compound of Formula (V) to mesylating reagent is 1:2. In one embodiment, the molar ratio of the compound of Formula (V) to mesylating reagent is about 1:1.2.
  • step 3.0 occurs in the presence of a base. In some embodiments, step 3.0 occurs in the presence of a nitrogen containing base. In some embodiments, step 3.0 occurs in the presence of NH 4 OH, triethylamine, diisopropylethylamine (DIEA or DIPEA), pyridine, lutidine, 4-dimethylaminopyridine, imidazole, or 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU). In one embodiment, the base is triethylamine (TEA).
  • the molar ratio of the compound of Formula (V) to base in step 3.0 is from about 1:1 to about 1:4. In one embodiment, the molar ratio of the compound of Formula (V) to base is about 1:4. In one embodiment, the molar ratio of the compound of Formula (V) to base is about 1:1.3.
  • Step 3.0 may occur in a solvent suitable for the reaction.
  • the solvent is an organic solvent or a mixture of organic solvents.
  • step 3.0 occurs in a solvent of dichloromethane (DCM).
  • step 3.0 occurs in a solvent of 2-methyl THF.
  • the volume of solvent in step 3.0 is from about 4 vol to about 20 vol. In one embodiment, the volume of the solvent in step 3.0 is about 20 vol. In one embodiment, the volume of the solvent in step 3.0 is about 15 vol. In one embodiment, the volume of the solvent in step 3.0 is about 10 vol.
  • step 3.0 occurs at a reaction temperature of less than 5° C. In some embodiments, step 3.0 occurs at a reaction temperature of from about 0° C. to about 5° C. In one embodiment, the reaction temperature is about 0° C. In one embodiment, the reaction temperature is about 5° C.
  • step 3.0 occurs at a reaction time from about 10 minutes to about 2 hours. In one embodiment, the reaction time is about 30 minutes.
  • the mesylating reagent in step 3.0 is methanesulfonic anhydride (Ms 2 O) and the base is triethylamine.
  • the molar ratios of the compound of Formula (V) to mesylating reagent and base are 1.2 and 1.3, respectively.
  • step 3.0 occurs in a solvent of DCM and a solvent volume of 10 vol. at 0° C. to room temperature.
  • step 3.0 occurs in a solvent of DCM and a solvent volume of 10 vol. at room temperature.
  • step 3.0 occurs in a solvent of 2-methyl THF and a solvent volume of 10 vol. at room temperature.
  • the compound of Formula (V) is purified by an aqueous extraction and/or precipitation from ethyl acetate with added heptane. In one embodiment, the compound of Formula (V) is purified by an aqueous extraction and/or precipitation from DCM with added heptane.
  • step 3.0 proceeds to greater than 90%, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or greater than 99% conversion within about 1 hour, as determined by HPLC and/or NMR. In some embodiments, step 3.0 provides less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% of an impurity distinct from the compound of Formula (III).
  • Impurities provided in step 3.0 may include, but are not limited to, starting materials. In some embodiments, the exemplified impurities observed in this step are elimination impurity (SP-3) and the starting material. In one embodiment, the total amount of impurities provided in step 3.0 is less than less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1%.
  • step 3.0 further comprises purification of the compound of Formula (III).
  • the compound of Formula (III) produced in step 3.0 is purified by aqueous extraction, and/or precipitation from a solvent by an anti-solvent.
  • the compound of Formula (III) is purified by aqueous extraction with dilute sodium bicarbonate.
  • extraction with sodium bicarbonate maintains the pH of the reaction mixture at about pH 7 or above.
  • extraction with sodium bicarbonate quenches resident methanesulfonic anhydride.
  • the compound of Formula (III) is precipitated from an organic solvent. In one embodiment, the compound of Formula (III) is precipitated from ethyl acetate. In one embodiment, the compound of Formula (III) is precipitated from DCM. In one embodiment, the compound of Formula (III) is precipitated from a solvent by addition of an anti-solvent. In one embodiment, the anti-solvent is heptane. In one embodiment, the final volume ratio of solvent to anti-solvent is from about 1:1 to about 1:4. In one embodiment, the final volume ratio of solvent to anti-solvent is about 1:3.
  • step 3.0 provides a compound of Formula (III) in a substantially pure form. In certain embodiments, step 3.0 provides a compound of Formula (III) in a substantially chemically pure form. In certain embodiments, step 3.0 provides a compound of Formula (III) substantially free of impurities. In certain embodiments, step 3.0 provides a compound of Formula (III) in a substantially enantiomerically pure form. In certain embodiments, step 3.0 provides a compound of Formula (III) in a solid form having a desired morphology or advantageous rheological properties.
  • a process for preparing a compound of Formula (V), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof comprising:
  • step 4.0 reacting a compound of Formula (VI):
  • the molar ratio of the compound of formula (VI) to the compound of Formula (VII) in step 4.0 is from about 1:1 to about 1:1.2. In one embodiment, the molar ratio of the compound of formula (VI) to the compound of Formula (VII) in step 4.0 is about 1:2. In one embodiment, the molar ratio of the compound of formula (VI) to the compound of Formula (VII) in step 4.0 is about 1:1.05. In one embodiment, the molar ratio of the compound of formula (VI) to the compound of Formula (VII) in step 4.0 is about 1:1.02.
  • step 4.0 occurs in the presence of a catalyst.
  • the catalyst is a palladium catalyst.
  • the palladium catalyst is Pd 2 (dba) 3 , Pd(PPh 3 ) 4 , PdCl 2 (PPh 3 ) 2 , PdCl 2 (PCy 3 ) 2 , PdCl 2 (dppf), PdCl 2 (dtbpf), or Pd(Amphos)Cl 2 .
  • the catalyst is PdCl 2 (dppf).
  • the catalyst is Pd(Amphos)Cl 2 .
  • the molar ratio of the compound of Formula (VI) to catalyst in step 4.0 is from about 1:0.001 (i.e. 0.1 mol %) to about 1:0.04 (i.e. 4 mol %). In some embodiments, the molar ratio of the compound of Formula (VI) to catalyst in step 4.0 is about 1:0.001, about 1:0.002, about 1:0.003, about 1:0.004, about 1:0.005, about 1:0.006, about 1:0.007, about 1:0.008, about 1:0.009, or about 1:0.01. In one embodiment, the molar ratio of the compound of Formula (VI) to catalyst in step 4.0 is about 1:0.04 (i.e. 4 mol %).
  • the molar ratio of the compound of Formula (VI) to catalyst in step 4.0 is about 1:0.005 (i.e. 0.5 mol %). In one embodiment, the molar ratio of the compound of Formula (VI) to catalyst in step 4.0 is about 1:0.002 (i.e. 0.2 mol %). In one embodiment, a catalyst loading of less than about 4 mol %, less than about 1 mol %, or about 0.5 mol %, is employed in step 4.0.
  • step 4.0 occurs in the presence of a base.
  • step 2.0 occurs in the presence of an alkali metal base.
  • the base is an alkali metal hydroxide, carbonate, hydrogencarbonate, phosphate, hydrogenphosphate, or dihydrogenphosphate.
  • the base is LiOH, NaOH, KOH, Na 2 CO 3 , K 2 CO 3 , Cs 2 CO 3 , NaHCO 3 , KHCO 3 , Na 3 PO 4 , K 3 PO 4 , Na 2 HPO 4 , K 2 HPO 4 , NaH 2 PO 4 , or KH 2 PO 4 .
  • the base is potassium carbonate (K 2 CO 3 ).
  • the base is potassium phosphate (K 3 PO 4 ).
  • the base is potassium phosphate trihydrate (K 3 PO 4 .3H 2 O).
  • the molar ratio of the compound of Formula (VI) to base in step 4.0 is from about 1:2 to about 1:4. In one embodiment, the molar ratio of the compound of Formula (VI) to base in step 4.0 is about 1:3. In one embodiment, the molar ratio of the compound of Formula (VI) to base in step 4.0 is about 1:3.05.
  • Step 4.0 may occur in a solvent suitable for the reaction.
  • the solvent is CPME, MeTHF, DME, DMAc, IPA, MeOH, DMF, DMA, NMP, ACN, DMSO, 1,4-dioxane, tetrahydrofuran, or water, or a mixture thereof.
  • the solvent is a mixture of an organic solvent and water.
  • step 4.0 occurs in a mixture of 1,4-dioxane and water.
  • step 4.0 occurs in a mixture of tetrahydrofuran and water.
  • step 4.0 occurs in a mixture of dimethyl formamide (DMF) and water.
  • DMF dimethyl formamide
  • step 4.0 occurs in a mixture of toluene and water.
  • the mixture an organic solvent and water has a ratio of organic solvent to water of from about 10:1 to about 3:1. In one embodiment, the ratio of organic solvent to water is about 10:1. In one embodiment, the ratio of organic solvent to water is about 5:1. In one embodiment, the mixture an organic solvent and water has a ratio of organic solvent to water of from about 1:2 to about 1:5. In one embodiment, the ratio of organic solvent to water is about 1:4.
  • the volume of solvent in step 4.0 is from about 2 vol to about 15 vol. In some embodiments, the volume of solvent in step 4.0 is from about 4 vol to about 15 vol. In one embodiment, the volume of the solvent in step 4.0 is about 11 vol. In one embodiment, the volume of the solvent in step 4.0 is about 6 vol. In one embodiment, the volume of the solvent in step 4.0 is about 2.5 vol.
  • step 4.0 occurs at a reaction temperature of from about 40° C. to about 90° C. In some embodiments, step 4.0 occurs at a reaction temperature of from about 40° C. to about 70° C. In some embodiments, step 4.0 occurs at a reaction temperature of from about 60° C. to about 70° C. In one embodiment, the reaction temperature is about 65° C. In one embodiment, the reaction temperature is about 50° C.
  • step 4.0 occurs at a reaction time from about 1 hour to about 4 hours. In one embodiment, the reaction time is from about 2 to about 3 hours. In one embodiment, the purity of the compound of Formula (V) provided in step 4.0 decreases with longer reaction times.
  • the molar ratio of the compound of formula (VI) to the compound of Formula (VII) in step 4.0 is about 1:1.2.
  • the catalyst is Pd(dppf)Cl 2 and the catalyst loading is 4 mol %.
  • the base is potassium carbonate (K 2 CO 3 ) and the molar ratio of the compound of Formula (VI) to potassium carbonate in step 4.0 is about 1:3.
  • the solvent in step 4.0 is an about 10:1 mixture of THF and water. In one embodiment, step 4.0 occurs at a reaction temperature of about 60 to about 65° C.
  • the molar ratio of the compound of formula (VI) to the compound of Formula (VII) in step 4.0 is about 1:1.05.
  • the catalyst is Pd(Amphos)Cl 2 and the catalyst loading is 0.05 mol %. In one embodiment, the catalyst is Pd(Amphos)Cl 2 and the catalyst loading is 0.5 mol %. In one embodiment, the catalyst is Pd(Amphos)Cl 2 and the catalyst loading is 0.4 mol %. In one embodiment, the catalyst is Pd(Amphos)Cl 2 and the catalyst loading is 0.3 mol %. In one embodiment, the catalyst is Pd(Amphos)Cl 2 and the catalyst loading is 0.2 mol %.
  • the base is potassium carbonate (K 2 CO 3 ) and the molar ratio of the compound of Formula (VI) to potassium carbonate in step 4.0 is about 1:3.
  • the solvent in step 4.0 is an about 5:1 mixture of dimethylformamide and water.
  • step 4.0 occurs at a reaction temperature of about 65° C. and a reaction time of about 2 to about 3 hours.
  • the molar ratio of the compound of formula (VI) to the compound of Formula (VII) in step 4.0 is about 1:1.02.
  • the catalyst is Pd(Amphos)Cl 2 and the catalyst loading is 0.2 mol %.
  • the base is potassium phosphate trihydrate (K 3 PO 4 .3H 2 O) and the molar ratio of the compound of Formula (VI) to potassium phosphate trihydrate in step 4.0 is about 1:3.05.
  • the solvent in step 4.0 is an about 1:4 mixture of toluene and water. In one embodiment, step 4.0 occurs at a reaction temperature of about 50° C.
  • step 4.0 proceeds to greater than 90%, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or greater than 99% conversion within about 2-3 hours, as determined by HPLC and/or NMR. In some embodiments, step 4.0 provides less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% of an impurity distinct from the compound of Formula (V). Impurities provided in step 4.0 may include, but are not limited to, starting materials and a compound of Formula (SP-6):
  • the total amount of impurities provided in step 3.0 is less than less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1%.
  • step 4.0 further comprises purification of the compound of Formula (V).
  • the compound of Formula (V) produced in step 4.0 is purified by aqueous extraction, palladium remediation, and/or precipitation from a solvent by an anti-solvent.
  • aqueous extraction comprises extraction of a composition comprising a compound of Formula (V) dissolved in an organic solvent with water.
  • the organic solvent is ethyl acetate.
  • aqueous extraction comprises extraction of a composition comprising a compound of Formula (V) dissolved in an organic solvent with an aqueous lithium chloride solution.
  • the aqueous lithium chloride solution is a 5% aqueous lithium chloride solution.
  • the aqueous lithium chloride solution is a 15% aqueous lithium chloride solution.
  • the organic solvent is ethyl acetate, DMF, MTBE, 2-MeTHF, or CPME, or a mixture thereof.
  • the organic solvent is a mixture of MTBE and DMF.
  • the organic solvent is MTBE.
  • the organic solvent is a mixture of MTBE and isopropyl ether.
  • the palladium remediation comprises treatment with any palladium scavenger known in the art.
  • the palladium scavenger is a thiopropyl silica scavenger.
  • the palladium remediation occurs at a temperature above room temperature, e.g., from about 40° C. to about 80° C., e.g., about 65° C.
  • the palladium remediation comprises treatment with a palladium scavenger for a period of about 1 hour to about 18 hours.
  • the palladium remediation comprises treatment with a palladium scavenger for a period of about 1 hour, about 3 hours, greater than about 3 hours, greater than 10 hours, greater than 14 hours, greater than 16 hours, or about 18 hours.
  • the palladium remediation comprises purifying the compound of Formula (V) by silica gel chromatography.
  • the palladium remediation comprises washing an organic solution of the compound of Formula (V) with aqueous L-cysteine.
  • the aqueous L-cysteine is 5 w % aqueous solution of L-cysteine.
  • the washing is conducted at elevated temperatures, (i.e. at a temperature above room temperature), for example at a temperature of about 35-50° C., or 40-45° C.
  • the palladium remediation comprises purifying the compound of Formula (V) by silica gel chromatography and washing an organic solution of the compound of Formula (V) with aqueous L-cysteine.
  • the palladium remediation comprises treating a solution of the compound of Formula (V) with activated carbon (e.g., via a CUNO filter).
  • the compound of Formula (V) is precipitated from an organic solvent. In one embodiment, the compound of Formula (V) is precipitated from MTBE. In one embodiment, the compound of Formula (V) is precipitated from a mixture of MTBE and isopropyl ether. In one embodiment, the compound of Formula (V) is precipitated from a solvent by addition of an anti-solvent. In one embodiment, the anti-solvent is isopropyl ether. In one embodiment, the anti-solvent is heptane. In one embodiment, the final ratio of solvent to anti-solvent is from about 1:1 to about 1:4. In one embodiment, the final ratio of solvent to anti-solvent is about 1:3.
  • the anti-solvent is added to the solvent at a temperature below 0° C.
  • the compound of Formula (V) is crystallized or recrystallized from an organic solvent or a mixture of organic solvents.
  • the compound of Formula (II) is crystallized or recrystallized from ethyl acetate, CPME, MTBE, toluene, or a mixture thereof, optionally by addition of an anti-solvent.
  • the anti-solvent is water.
  • the anti-solvent is a non-polar organic solvent.
  • the non-polar organic solvent is a hydrocarbon solvent.
  • the anti-solvent is heptane.
  • the solvent is ethyl acetate and the anti-solvent is heptane. In one embodiment, the solvent is toluene and the anti-solvent is heptane. In one embodiment, the final volume ratio of solvent to anti-solvent is from about 1:2 to about 1:6.
  • step 4.0 provides a compound of Formula (V) in a substantially pure form. In certain embodiments, step 4.0 provides a compound of Formula (V) in a substantially chemically pure form. In certain embodiments, step 4.0 provides a compound of Formula (V) substantially free of impurities. In certain embodiments, step 4.0 provides a compound of Formula (V) in a substantially enantiomerically pure form. In certain embodiments, step 4.0 provides a compound of Formula (V) in a solid form having a desired morphology or advantageous rheological properties.
  • a process for preparing a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof is prepared by a process comprising:
  • step 1.0 cyclizing a compound of Formula (II) or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, to provide a compound of Formula (I), or an enantiomer, a mixture of enantiomers, or a tautomer thereof, or a pharmaceutically acceptable salt thereof, wherein the compound of Formula (II) is prepared by a process comprising: (step 2.0) reacting a compound of Formula (III) or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, with a compound of Formula (IV), or a pharmaceutically acceptable salt thereof, wherein the compound of Formula (III) is prepared by a process comprising: (step 3.0) reacting a compound of Formula (V) or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, with a mesylating reagent; and wherein the compound of Formula (V) is prepared
  • the processes provided herein further comprise a step of providing the compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, in a solid form.
  • the solid form is a crystalline form.
  • provided herein is a crystalline form of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, prepared by the process.
  • composition comprising a compound of Formula (I):
  • Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, in the pharmaceutical composition is free base of Compound 1.
  • the free base of Compound 1 is amorphous.
  • the free base of Compound 1 is one of the solid forms of free base of Compound 1 provided herein.
  • the free base of Compound 1 is Form 1 of the free base of Compound 1.
  • Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, in the pharmaceutical composition is a pharmaceutically acceptable salt of Compound 1.
  • the salt is a chloride, phosphate, besylate, mesylate, citrate or maleate salt of Compound 1.
  • the salt is a besylate salt of Compound 1 provided herein.
  • the salt is a phosphate salt of Compound 1 provided herein.
  • the salt is amorphous.
  • the salt is one of the crystalline forms of a salt of Compound 1 provided herein.
  • the total weight of the pharmaceutical composition does not include coating of the pharmaceutical composition (e.g., an Opadry II coat of a tablet pharmaceutical composition provided herein).
  • the amount of Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, in the pharmaceutical composition is from about 1% to about 30% w/w. In one embodiment, the amount is from about 3% to about 20% w/w. In one embodiment, the amount is from about 5% to about 17% w/w. In one embodiment, the amount is from about 3% to about 7% w/w.
  • the amount is from about 4% to about 6% w/w. In one embodiment, the amount is about 3, about 4, about 5, about 6, or about 7% w/w. In one embodiment, the amount is about 5% w/w. In one embodiment, the amount is from about 15% to about 20% w/w. In one embodiment, the amount is from about 10% to about 17% w/w. In one embodiment, the amount is from about 15% to about 17% w/w. In one embodiment, the amount is from about 16% to about 18% w/w. In one embodiment, the amount is from about 16% to about 17% w/w. In one embodiment, the amount is about 15, about 16, about 17, about 18, about 19, or about 20% w/w. In one embodiment, the amount is about 17% w/w. In one embodiment, the amount is about 16.7% w/w. In one embodiment, the amount is about 16.67% w/w.
  • the diluent is mannitol.
  • the diluent is Pearlitol 100SD, Pearlitol 110 C, Pearlitol 160 C, Pearlitol 25 C, Pearlitol 300 DC, Pearlitol 400 DC, Pearlitol 500 DC, Pearlitol SW-F 200, Parteck M 100, Parteck M 200, or lactose monohydrate.
  • the diluent can be a saccharide, e.g., selected from the group consisting of sucrose, dextrose, lactose, fructose, mannitol, a cellulosic derivative, calcium phosphate, calcium carbonate, and a mixture thereof.
  • the diluent can be mannitol or microcrystalline cellulose (e.g., SMCC 90). In one embodiment, the diluent is mannitol Pearlitol 50C. In one embodiment, the diluent is mannitol Pearlitol 200SD.
  • the amount of the diluent in the pharmaceutical composition is from about 50% to about 95% w/w. In one embodiment, the amount is from about 60% to about 90% w/w. In one embodiment, the amount is from about 65% to about 87% w/w. In one embodiment, the amount is from about 62% to about 68% w/w. In one embodiment, the amount is from about 64% to about 66% w/w. In one embodiment, the amount is about 62, about 63, about 64, about 65, about 66, about 67, or about 68% w/w. In one embodiment, the amount is about 65% w/w. In one embodiment, the amount is from about 85% to about 90% w/w.
  • the amount is from about 86% to about 88% w/w. In one embodiment, the amount is about 85, about 86, about 87, about 88, about 89, or about 90% w/w. In one embodiment, the amount is about 87% w/w.
  • the diluent is mannitol, startch, SMCC, lactose, dibasic calcium phosphate (DCP). In some embodiments, the diluent is mannitol. In some embodiments, the diluent is SMCC. In some embodiments, the diluent is a mixture of mannitol and SMCC.
  • the weight ratio of mannitol to SMCC is in a range of about 1:5 to about 35:1. In some embodiments, the weight ratio of mannitol to SMCC is in a range of about 1:1 to about 35:1. In some embodiments, the weight ratio of mannitol to SMCC is in a range of about 3:1 to about 30:1. In some embodiments, the weight ratio of mannitol to SMCC is in a range of about 6:1 to about 1:1. In some embodiments, the weight ratio of mannitol to SMCC is in a range of about 1:5 to about 1:1. In some embodiment, the weight ratio of mannitol to SMCC is about 5:1. In some embodiment, the weight ratio of mannitol to SMCC is about 5.3:1.
  • Silicified microcrystalline cellulose is a unique combination of microcrystalline cellulose (MCC) and colloidal silicon dioxide (CSD) as a high functionality and multifunctional excipient
  • MCC microcrystalline cellulose
  • CSS colloidal silicon dioxide
  • SMCC is unique in that it imparts both optimum compaction and superior flow to formulations as a diluent. It also exhibits both brittle fracture and plastic deformation characteristics, leading to good binding properties.
  • the binder is hydroxypropyl cellulose (HPC), silicified microcrystalline cellulose (SMCC), or a mixture thereof.
  • the binder is HPC.
  • the binder is SMCC. In one embodiment, the binder is a mixture of HPC and SMCC. In one embodiment, the weight ratio of HPC to SMCC is from about 1:8 to about 1:14. In one embodiment, the weight ratio of HPC to SMCC is about 1:8, about 1:9, about 1:10, about 1:11, about 1:12, about 1:13, or about 1:14. In one embodiment, the weight ratio of HPC to SMCC is about 1:11. In one embodiment, the amount of HPC in the pharmaceutical composition is about 1.1% w/w. In one embodiment, the amount of SMCC in the pharmaceutical composition is about 12.2% w/w.
  • the binder is carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia, or a mixture thereof.
  • the binder is one or more polysaccharides and their derivatives such as starches, cellulose or modified cellulose such as MCC and cellulose ethers such as hydroxypropyl cellulose (HPC); sodium alginate, gelatin, polyvinyl pyrrolidone (PVP), methylcellulose, hydroxy propyl methyl cellulose (HPMC), polymethacrylates, sodium carboxy methyl cellulose, polyethylene glycol (PEG), or a mixture thereof.
  • HPC hydroxypropyl cellulose
  • PVP polyvinyl pyrrolidone
  • HPMC hydroxy propyl methyl cellulose
  • PEG polyethylene glycol
  • the amount of the binder in the pharmaceutical composition is from about 1% to about 20% w/w. In one embodiment, the amount is from about 2% to about 16% w/w. In one embodiment, the amount is from about 2% to about 14% w/w. In one embodiment, the amount is from about 4% to about 14% w/w. In one embodiment, the amount is from about 2% to about 6% w/w. In one embodiment, the amount is from about 2% to about 5% w/w. In one embodiment, the amount is from about 3% to about 5% w/w. In one embodiment, the amount is about 2, about 3, about 4, about 5, or about 6% w/w. In one embodiment, the amount is about 4% w/w.
  • the amount is from about 12% to about 16% w/w. In one embodiment, the amount is from about 13% to about 15% w/w. In one embodiment, the amount is about 12, about 13, about 14, about 15, or about 16% w/w. In one embodiment, the amount is about 14% w/w. In one embodiment, the amount is about 13.3% w/w. In one embodiment, the amount is about 1, about 1.1, about 1.2, about 1.3, about 1.4, about 1.5, about 1.6, about 1.7, about 1.8, about 1.9, or about 2% w/w. In one embodiment, the amount of binder is about 1.10% w/w.
  • the disintegrant is sodium starch glycolate (SSG). In one embodiment, the disintegrant is cross-linked sodium carboxymethyl cellulose. In one embodiment, the disintegrant is one or more selected from the group consisting of cross-linked sodium carboxymethylcellulose, starch, sodium starch glycolate, cross-linked polyvinylpyrrolidone, crospovidone and a mixture thereof.
  • the amount of the disintegrant in the pharmaceutical composition is from about 1% to about 8% w/w. In one embodiment, the amount is from about 2% to about 5% w/w. In one embodiment, the amount is from about 2% to about 4% w/w. In one embodiment, the amount is from about 2.9% to about 4% w/w.
  • the amount is about 2, about 2.1, about 2.2, about 2.3, about 2.4, about 2.5, about 2.6, about 2.7, about 2.8, about 2.9, about 3, about 3.1, about 3.2, about 3.3, about 3.4, about 3.5, about 3.6, about 3.7, about 3.8, about 3.9, about 4, about 4.1, about 4.2, about 4.3, about 4.4, about 4.5, about 4.6, about 4.7, about 4.8, about 4.9, or about 5% w/w.
  • the amount is about 2.9% w/w.
  • the amount is about 3.9% w/w.
  • the amount is about 4% w/w.
  • the lubricant is magnesium stearate. In another embodiment, the lubricant is sodium lauryl sulfate. In one embodiment, the lubricant is one or more metallic salts of fatty acids such as magnesium stearate and steric acid, Carbowax (PEG) 4000/6000, sodium oleate, sterotex, sodium benzoate, Talc, sodium acetate, waxes, sodium lauryl sulfate, Stear-O-Wet, Mg-lauryl sulfate, and glyceryl behenate (Compritol 888).
  • fatty acids such as magnesium stearate and steric acid, Carbowax (PEG) 4000/6000, sodium oleate, sterotex, sodium benzoate, Talc, sodium acetate, waxes, sodium lauryl sulfate, Stear-O-Wet, Mg-lauryl sulfate, and glyceryl behenate (Compri
  • the amount of the lubricant in the pharmaceutical composition is from about 0.10% to about 3% w/w. In one embodiment, the amount is from about 0.8% to about 1.4% w/w. In one embodiment, the amount is from about 1% to about 1.2% w/w. In one embodiment, the amount is about 0.8, about 0.9, about 1, about 1.1, about 1.2, about 1.3, or about 1.4% w/w. In one embodiment, the amount is about 10% w/w. In one embodiment, the amount is about 1.10% w/w. In one embodiment, the amount is about 1.2% w/w.
  • the pharmaceutical composition is a tablet comprising: Compound 1 at an amount of from about 4.0% to about 5.5% w/w of the total weight of the pharmaceutical composition; mannitol at an amount of from about 80% to about 88% w/w of the total weight of the pharmaceutical composition; silicified microcrystalline cellulose at an amount of from about 3.0% to about 4.0% w/w of the total weight of the pharmaceutical composition; hydroxypropyl cellulose at an amount of from about 0.2% to about 0.4% w/w of the total weight of the pharmaceutical composition; sodium starch glycolate at an amount of from about 2.2% to about 3.5% w/w of the total weight of the pharmaceutical composition; and magnesium stearate at an amount of from about 0.5% to about 1.5% w/w of the total weight of the pharmaceutical composition.
  • the tablet composition comprises: Compound 1 at an amount of about 5% w/w of the total weight of the pharmaceutical composition; mannitol at an amount of about 84% w/w of the total weight of the pharmaceutical composition; silicified microcrystalline cellulose at an amount of about 3.5% w/w of the total weight of the pharmaceutical composition; hydroxypropyl cellulose at an amount of about 0.3% w/w of the total weight of the pharmaceutical composition; sodium starch glycolate at an amount of about 2.8% w/w of the total weight of the pharmaceutical composition; and magnesium stearate at an amount of about 1.0% w/w of the total weight of the pharmaceutical composition.
  • the tablet composition has a total weight of about 100 mg with about 5 mg of a solid form (e.g. Form 1) of Compound 1.
  • the pharmaceutical composition is a tablet comprising: Compound 1 at an amount of from about 15.0% to about 17.5% w/w of the total weight of the pharmaceutical composition; mannitol at an amount of from about 60% to about 66% w/w of the total weight of the pharmaceutical composition; silicified microcrystalline cellulose at an amount of from about 10% to about 13% w/w of the total weight of the pharmaceutical composition; hydroxypropyl cellulose at an amount of from about 0.5 to about 1.5% w/w of the total weight of the pharmaceutical composition; sodium starch glycolate at an amount of from about 3.0% to about 4.5% w/w of the total weight of the pharmaceutical composition; and magnesium stearate at an amount of from about 0.8 to about 1.5% w/w of the total weight of the pharmaceutical composition.
  • the tablet composition comprises: Compound 1 at an amount of about 16.67% w/w of the total weight of the pharmaceutical composition; mannitol at an amount of about 62% w/w of the total weight of the pharmaceutical composition; silicified microcrystalline cellulose at an amount of about 11.7% w/w of the total weight of the pharmaceutical composition; hydroxypropyl cellulose at an amount of about 1.0% w/w of the total weight of the pharmaceutical composition; sodium starch glycolate at an amount of about 3.7% w/w of the total weight of the pharmaceutical composition; and magnesium stearate at an amount of about 1.1% w/w of the total weight of the pharmaceutical composition.
  • the tablet composition has a total weight of about 150 mg.
  • the tablet composition has a total weight of about 150 mg with about 25 mg of a solid form (e.g. Form 1) of Compound 1.
  • the tablet composition comprises: Compound 1 at an amount of about 16.7% w/w of the total weight of the pharmaceutical composition; mannitol at an amount of about 65% w/w of the total weight of the pharmaceutical composition; silicified microcrystalline cellulose at an amount of about 12.2% w/w of the total weight of the pharmaceutical composition; hydroxypropyl cellulose at an amount of about 1.1% w/w of the total weight of the pharmaceutical composition; sodium starch glycolate at an amount of about 3.9% w/w of the total weight of the pharmaceutical composition; and magnesium stearate at an amount of about 1.1% w/w of the total weight of the pharmaceutical composition.
  • the tablet composition comprises: Compound 1 at an amount of about 16.67% w/w of the total weight of the pharmaceutical composition; mannitol at an amount of about 64.97% w/w of the total weight of the pharmaceutical composition; silicified microcrystalline cellulose at an amount of about 12.22% w/w of the total weight of the pharmaceutical composition; hydroxypropyl cellulose at an amount of about 1.11% w/w of the total weight of the pharmaceutical composition; sodium starch glycolate at an amount of about 3.89% w/w of the total weight of the pharmaceutical composition; and magnesium stearate at an amount of about 1.14% w/w of the total weight of the pharmaceutical composition.
  • the tablet composition has a total weight of about 150 mg.
  • the tablet composition has a total weight of about 150 mg with about 25 mg of a solid form (e.g. Form 1) of Compound 1. In one embodiment, the tablet composition has a total weight of about 300 mg. In one embodiment, the tablet composition has a total weight of about 300 mg with about 50 mg of a solid form (e.g. Form 1) of Compound 1. In one embodiment, the tablet composition has a total weight of about 450 mg. In one embodiment, the tablet composition has a total weight of about 450 mg with about 75 mg of a solid form (e.g. Form 1) of Compound 1. In one embodiment, the tablet composition has a total weight of about 600 mg. In one embodiment, the tablet composition has a total weight of about 600 mg with about 100 mg of a solid form (e.g. Form 1) of Compound 1.
  • the pharmaceutical compositions may conveniently be presented in unit dosage form.
  • the unit dosage form is a tablet.
  • the unit dosage form is a tablet of 5 mg (by weight of free base Compound 1) dose strength.
  • the unit dosage form is a tablet of 25 mg (by weight of free base Compound 1) dose strength.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, the particular mode of administration.
  • the pharmaceutical composition provided herein comprises (i) a granular formulation comprising Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, (ii) extragranular diluent, (iii) extragranular disintegrant, and (iv) extragranular lubricant.
  • the amount of the granular formulation in the pharmaceutical composition is from about 10% to about 65% w/w. In one embodiment, the amount is from about 13% to about 60% w/w. In one embodiment, the amount is from about 16% to about 56% w/w. In one embodiment, the amount is from about 13% to about 20% w/w. In one embodiment, the amount is from about 15% to about 18% w/w. In one embodiment, the amount is about 13, about 14, about 15, about 16, about 17, about 18, about 19, or about 20% w/w. In one embodiment, the amount is about 16% w/w. In one embodiment, the amount is about 17% w/w. In one embodiment, the amount is about 16.7% w/w.
  • the amount is about 16.67% w/w. In one embodiment, the amount is from about 50% to about 60% w/w. In one embodiment, the amount is from about 54% to about 57% w/w. In one embodiment, the amount is about 50, about 51, about 52, about 53, about 54, about 55, about 56, about 57, about 58, about 59, or about 60% w/w. In one embodiment, the amount is about 55% w/w. In one embodiment, the amount is about 56% w/w. In one embodiment, the amount is about 55.6% w/w. In one embodiment, the amount is about 55.55% w/w.
  • the amount of the extragranular diluent in the pharmaceutical composition is from about 30% to about 85% w/w. In one embodiment, the amount is from about 35% to about 85% w/w. In one embodiment, the amount is from about 40% to about 80% w/w. In one embodiment, the amount is from about 35% to about 45% w/w. In one embodiment, the amount is from about 40% to about 42% w/w. In one embodiment, the amount is about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, or about 45% w/w. In one embodiment, the amount is about 410% w/w. In one embodiment, the amount is about 40.95% w/w.
  • the amount is from about 75% to about 85% w/w. In one embodiment, the amount is from about 79% to about 81% w/w. In one embodiment, the amount is about 75, about 76, about 77, about 78, about 79, about 80, about 81, about 82, about 83, about 84, or about 85% w/w. In one embodiment, the amount is about 80% w/w. In one embodiment, the amount is about 79.8% w/w. In one embodiment, the amount is about 79.83% w/w.
  • the amount of the extragranular disintegrant in the pharmaceutical composition is from about 1% to about 4% w/w. In one embodiment, the amount is from about 2% to about 3% w/w. In one embodiment, the amount is about 2, about 2.1, about 2.2, about 2.3, about 2.4, about 2.5, about 2.6, about 2.7, about 2.8, about 2.9, or about 3% w/w. In one embodiment, the amount is about 2.5% w/w.
  • the amount of the extragranular lubricant in the pharmaceutical composition is from about 0.1% to about 2.5% w/w. In one embodiment, the amount is from about 0.5% to about 1.5% w/w. In one embodiment, the amount is about 0.5, about 0.6, about 0.7, about 0.8, about 0.9, about 1, about 1.1, about 1.2, about 1.3, about 1.4, or about 1.5% w/w. In one embodiment, the amount is about 1% w/w.
  • the granular formulation comprising Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, the binder, intragranular diluent which is optional, intragranular disintegrant, and intragranular lubricant.
  • the granular formulation comprising a solid form (e.g. Form 1) of Compound 1.
  • the amount of Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, in the granular formulation is from about 20% to about 80% w/w. In one embodiment, the amount is from about 30% to about 70% w/w. In one embodiment, the amount is from about 30% to about 50% w/w. In one embodiment, the amount is from about 30% to about 40% w/w. In one embodiment, the amount is about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, or about 80% w/w. In one embodiment, the amount is about 30% w/w. In one embodiment, the amount is about 40% w/w. In one embodiment, the amount is about 50% w/w.
  • the amount of the binder in the granular formulation is from about 15% to about 35% w/w. In one embodiment, the amount is from about 20% to about 30% w/w. In one embodiment, the amount is about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, or about 30% w/w. In one embodiment, the amount is about 24% w/w.
  • the amount of the intragranular disintegrant in the granular formulation is from about 1% to about 4% w/w. In one embodiment, the amount is from about 2% to about 3% w/w. In one embodiment, the amount is about 2, about 2.1, about 2.2, about 2.3, about 2.4, about 2.5, about 2.6, about 2.7, about 2.8, about 2.9, or about 3% w/w. In one embodiment, the amount is about 2.5% w/w.
  • the amount of the intragranular lubricant in the granular formulation is from about 0.1% to about 0.5% w/w. In one embodiment, the amount is about 0.1, about 0.15, about 0.2, about 0.25, about 0.3, about 0.35, about 0.4, about 0.45, or about 0.5% w/w. In one embodiment, the amount is about 0.25% w/w.
  • the amount of the intragranular diluent in the granular formulation is from about 0% to about 60% w/w. In one embodiment, the amount is from about 35% to about 50% w/w. In one embodiment, the amount is from about 40% to about 45% w/w. In one embodiment, the amount is about 35, about 36, about 37, about 38, about 39, about 40, about 41, about 42, about 43, about 44, about 45, about 46, about 47, about 48, about 49, or about 50% w/w. In one embodiment, the amount is about 43% w/w. In one embodiment, the amount is about 43.2% w/w. In one embodiment, the amount is about 43.3% w/w. In one embodiment, the amount is about 43.25% w/w. In one embodiment, the amount of the intragranular diluent in the granular formulation is the balance of the granular formulation.
  • the pharmaceutical composition comprises: (i) a granular formulation at an amount of from about 16% to about 17.5% w/w of the total weight of the pharmaceutical composition; (ii) mannitol at an amount of from about 79% to about 80.5% w/w of the total weight of the pharmaceutical composition; (iii) sodium starch glycolate at an amount of about 2.5% w/w of the total weight of the pharmaceutical composition; and (iv) magnesium stearate at an amount of about 1% w/w of the total weight of the pharmaceutical composition; wherein the granular formulation comprises: (i.a) Compound 1 at an amount of from about 30% to about 30.5% w/w in the granular formulation; (i.b) mannitol at an amount of from about 42.75% to about 43.25% w/w of the total weight of the granular formulation; (i.c) silicified microcrystalline cellulose at an amount of about 22% w/w of the total weight of the granular formulation; (i.d
  • the pharmaceutical composition comprises: (i) a granular formulation at an amount of about 16.7% (in one embodiment, about 16.67%) w/w of the total weight of the pharmaceutical composition; (ii) mannitol at an amount of about 79.8% (in one embodiment, about 79.83%) w/w of the total weight of the pharmaceutical composition; (iii) sodium starch glycolate at an amount of about 2.5% w/w of the total weight of the pharmaceutical composition; and (iv) magnesium stearate at an amount of about 1% w/w of the total weight of the pharmaceutical composition; wherein the granular formulation comprises: (i.a) Compound 1 at an amount of about 30% w/w in the granular formulation; (i.b) mannitol at an amount of about 43.25% w/w of the total weight of the granular formulation; (i.c) silicified microcrystalline cellulose at an amount of about 22% w/w of the total weight of the granular formulation; (i.d)
  • the pharmaceutical composition comprises: (i) a granular formulation at the amount of from about 55% to about 56.5% w/w of the total weight of the pharmaceutical composition; (ii) mannitol at the amount of from about 40% to about 41.5% w/w of the total weight of the pharmaceutical composition; (iii) sodium starch glycolate at the amount of about 2.5% w/w of the total weight of the pharmaceutical composition; and (iv) magnesium stearate at the amount of about 1% w/w of the total weight of the pharmaceutical composition; wherein the granular formulation comprises: (i.a) Compound 1 at an amount of from about 30% to about 30.5% w/w in the granular formulation; (i.b) mannitol at an amount of from about 42.75% to about 43.25% w/w of the total weight of the granular formulation; (i.c) silicified microcrystalline cellulose at an amount of about 22% w/w of the total weight of the granular formulation; (i.d
  • the pharmaceutical composition comprises: (i) a granular formulation at the amount of about 55.6% w/w of the total weight of the pharmaceutical composition; (ii) mannitol at the amount of about 40.9% w/w of the total weight of the pharmaceutical composition; (iii) sodium starch glycolate at the amount of about 2.5% w/w of the total weight of the pharmaceutical composition; and (iv) magnesium stearate at the amount of about 1% w/w of the total weight of the pharmaceutical composition; wherein the granular formulation comprises: (i.a) Compound 1 at an amount of about 30% w/w in the granular formulation; (i.b) mannitol at an amount of about 43.25% w/w of the total weight of the granular formulation; (i.c) silicified microcrystalline cellulose at an amount of about 22% w/w of the total weight of the granular formulation; (i.d) hydroxypropyl cellulose at an amount of about 2% w/w of the
  • the pharmaceutical composition provided herein is an oral dosage form.
  • the oral dosage form is a tablet.
  • the oral dosage form is an immediate release tablet.
  • the pharmaceutical composition is prepared by a process comprising blending a granule formulation comprising Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, with one or more extragranular excipient(s).
  • the one or more extragranular excipient(s) comprises an extragranular diluent (e.g., mannitol), an extragranular disintegrant (e.g., sodium starch glycolate), and an extragranular lubricant (e.g., magnesium stearate).
  • the extragranular lubricant e.g., magnesium stearate
  • the extragranular lubricant is blended after the other components are blended first.
  • the granule formulation is prepared by a process feeding an intragranular blend comprising Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, through a roller compactor and optionally a comil to provide the granule formulation.
  • the intragranular blend is prepared by a process comprising blending Compound 1, or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, with one or more intragranular excipient(s).
  • the one or more intragranular excipient(s) comprises an intragranular diluent (e.g., mannitol), an intragranular binder (e.g., a mixture of silicified microcrystalline cellulose and hydroxypropyl cellulose), an intragranular disintegrant (e.g., sodium starch glycolate), and an intragranular lubricant (e.g., magnesium stearate).
  • an intragranular lubricant e.g., magnesium stearate
  • the intragranular lubricant is blended after the other components are blended first.
  • the pharmaceutical composition provided herein has a particle size of between about 10 ⁇ m and about 500 ⁇ m. In one embodiment, the particle size is between about 10 ⁇ m and about 50 ⁇ m. In one embodiment, the particle size is between about 25 ⁇ m and about 35 ⁇ m. In one embodiment, the particle size is between about 50 ⁇ m and about 150 ⁇ m. In one embodiment, the particle size is between about 120 ⁇ m and about 140 ⁇ m. In one embodiment, the particle size is between about 150 ⁇ m and about 400 ⁇ m. In one embodiment, the particle size is between about 320 ⁇ m and about 360 ⁇ m.
  • the pharmaceutical composition provided herein comprises a population of particles having an average particle size of between about 10 ⁇ m and about 150 ⁇ m.
  • the average particle size (e.g., D10) is between about 10 ⁇ m and about 50 ⁇ m. In one embodiment, the average particle size (e.g., D10) is between about 10 ⁇ m and about 30 ⁇ m. In one embodiment, the average particle size (e.g., D10) is between about 10 ⁇ m and about 20 ⁇ m. In one embodiment, the average particle size (e.g., D50) is between about 50 ⁇ m and about 100 ⁇ m. In one embodiment, the average particle size (e.g., D50) is between about 50 ⁇ m and about 100 ⁇ m.
  • the average particle size (e.g., D50) is between about 60 ⁇ m and about 90 ⁇ m. In one embodiment, the average particle size (e.g., D50) is between about 70 ⁇ m and about 80 ⁇ m. In one embodiment, the average particle size (e.g., D90) is between about 100 ⁇ m and about 150 ⁇ m. In one embodiment, the average particle size (e.g., D90) is between about 110 ⁇ m and about 150 ⁇ m. In one embodiment, the average particle size (e.g., D90) is between about 110 ⁇ m and about 120 ⁇ m.
  • the average particle size (e.g., D10) is about 10 ⁇ m, about 11 ⁇ m, about 12 ⁇ m, about 13 ⁇ m, about 14 ⁇ m, about 15 ⁇ m, about 16 ⁇ m, about 17 ⁇ m, about 18 ⁇ m, about 19 ⁇ m, or about 20 ⁇ m.
  • the average particle size (e.g., D50) is about 50 ⁇ m, about 55 ⁇ m, about 60 ⁇ m, about 65 ⁇ m, about 70 ⁇ m, about 75 ⁇ m, about 80 ⁇ m, about 85 ⁇ m, about 90 ⁇ m, about 95 ⁇ m, or about 100 ⁇ m.
  • the average particle size (e.g., D90) is about 110 ⁇ m, about 115 ⁇ m, about 120 ⁇ m, about 125 ⁇ m, about 130 ⁇ m, about 135 ⁇ m, about 140 ⁇ m, about 145 ⁇ m, or about 150 ⁇ m.
  • the particles having the particle size or the population of particles having the average particle size is obtained by milling a solid form of Compound 1 (e.g. Form 1).
  • the milling is wet milling.
  • the wet milling is conducted according to means described herein or known in the art.
  • the milling is dry milling.
  • the dry milling is conducted according to means described herein or known in the art.
  • the particle size or average particle size is determined using any means known in the art, for example using a Malvern Mastersizer 3000 or analogous instrument.
  • the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has a chemical and/or physical purity of at least about 90%.
  • the chemical and/or physical purity of Compound 1 in a pharmaceutical composition is the amount of Compound 1 detected in a representative sample of the pharmaceutical composition (e.g., the amount detected by HPLC or other analytical technique) divided by the theoretical amount of Compound 1 in the formulation.
  • the pharmaceutical composition comprising Compound 1 has a chemical and/or physical purity of at least about 95%.
  • the pharmaceutical composition comprising Compound 1 has a chemical and/or physical purity of at least about 96%.
  • the pharmaceutical composition comprising Compound 1 has a chemical and/or physical purity of at least about 97%. In certain embodiments, the pharmaceutical composition comprising Compound 1 has a chemical and/or physical purity of at least about 98%. In certain embodiments, the pharmaceutical composition comprising Compound 1 has a chemical and/or physical purity of at least about 99%. In certain embodiments, the pharmaceutical composition comprising Compound 1 has a chemical and/or physical purity of about 99.4%. In certain embodiments, the pharmaceutical composition comprising Compound 1 has a chemical and/or physical purity of about 101.1%. In certain embodiments, the pharmaceutical composition comprising Compound 1 has an enantiomeric purity of at least about 99.5%.
  • the pharmaceutical composition comprising Compound 1 has an enantiomeric purity of about 100%. In certain embodiments, the pharmaceutical composition comprising Compound 1 has a chemical, physical, and/or enantiomeric purity of at least about 95% over 12 months.
  • the chemical and/or physical purity of the Compound 1 in the pharmaceutical composition is at least about 90 wt %. In certain embodiments, the chemical and/or physical purity of the Compound 1 in the pharmaceutical composition is at least about 95 wt %. In certain embodiments, the chemical and/or physical purity of the Compound 1 in the pharmaceutical composition is at least about 96 wt %. In certain embodiments, the chemical and/or physical purity of the Compound 1 in the pharmaceutical composition is at least about 97 wt %.
  • the chemical and/or physical purity of the Compound 1 in the pharmaceutical composition is at least about 98 wt %. In certain embodiments, the chemical and/or physical purity of the Compound 1 in the pharmaceutical composition is at least about 99 wt %. In certain embodiments, the chemical and/or physical purity of the Compound 1 in the pharmaceutical composition is about 99.4 wt %. In certain embodiments, the chemical and/or physical purity of the Compound 1 in the pharmaceutical composition is about 101.1 wt %. In certain embodiments, the enantiomeric purity of the Compound 1 in the pharmaceutical composition is at least about 99.5 wt %. In certain embodiments, the enantiomeric purity of the Compound 1 in the pharmaceutical composition is about 100 wt %.
  • the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 3% by area of degradation products. In certain embodiments, the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 2% by area of degradation products. In certain embodiments, the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 1% by area of degradation products. In certain embodiments, the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 0.5% by area of degradation products.
  • the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 0.1% by area of degradation products. In certain embodiments, the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has about 0.1% by area of degradation products.
  • the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 0.5% by area of one or more degradation product, respectively. In certain embodiments, the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 0.4% by area of one or more degradation product, respectively. In certain embodiments, the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 0.3% by area of one or more degradation product, respectively. In certain embodiments, the pharmaceutical composition comprising Compound 1 (e.g.
  • the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 0.2% by area of one or more degradation product, respectively.
  • the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 0.1% by area of one or more degradation product, respectively.
  • the pharmaceutical composition comprising Compound 1 (e.g. in a solid dosage form such as a tablet comprising Compound 1) has less than about 0.05% by area of one or more degradation product, respectively.
  • a pharmaceutical preparation suitable for use in a human patient comprising a compound provided herein (e.g., a compound of Formula (I), and one or more pharmaceutically acceptable excipients.
  • the pharmaceutical preparations may be for use in treating or preventing a condition or disease as described herein.
  • a compound provided herein may be used in the manufacture of medicaments for the treatment of any diseases or conditions disclosed herein.
  • compositions and methods provided herein may be utilized to treat a subject in need thereof.
  • the subject is a mammal such as a human, or a non-human mammal.
  • the composition or the compound is preferably administered as a pharmaceutical composition comprising, for example, a compound provided herein and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are well known in the art and include, for example, aqueous solutions such as water or physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, oils such as olive oil, or injectable organic esters.
  • the aqueous solution is pyrogen-free, or substantially pyrogen-free.
  • the excipients can be chosen, for example, to effect delayed release of an agent or to selectively target one or more cells, tissues or organs.
  • the pharmaceutical composition can be in dosage unit form such as tablet, capsule (including sprinkle capsule and gelatin capsule), granule, lyophile for reconstitution, powder, solution, syrup, suppository, injection or the like.
  • the composition can also be present in a transdermal delivery system, e.g., a skin patch.
  • the composition can also be present in a solution suitable for topical administration, such as an eye drop.
  • a pharmaceutically acceptable carrier can contain physiologically acceptable agents that act, for example, to stabilize, increase solubility or to increase the absorption of a compound such as a compound provided herein.
  • physiologically acceptable agents include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients.
  • the choice of a pharmaceutically acceptable carrier, including a physiologically acceptable agent depends, for example, on the route of administration of the composition.
  • the preparation or pharmaceutical composition can be a self-emulsifying drug delivery system or a self-microemulsifying drug delivery system.
  • the pharmaceutical composition also can be a liposome or other polymer matrix, which can have incorporated therein, for example, a compound provided herein.
  • Liposomes for example, which comprise phospholipids or other lipids, are nontoxic, physiologically acceptable and metabolizable carriers that are relatively simple to make and administer.
  • phrases “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material.
  • a pharmaceutically acceptable material such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the subject.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide;
  • a pharmaceutical composition can be administered to a subject by any of a number of routes of administration including, for example, orally (for example, drenches as in aqueous or non-aqueous solutions or suspensions, tablets, capsules (including sprinkle capsules and gelatin capsules), boluses, powders, granules, pastes for application to the tongue); absorption through the oral mucosa (e.g., sublingually); anally, rectally or vaginally (for example, as a pessary, cream or foam); parenterally (including intramuscularly, intravenously, subcutaneously or intrathecally as, for example, a sterile solution or suspension); nasally; intraperitoneally; subcutaneously; transdermally (for example as a patch applied to the skin); and topically (for example, as a cream, ointment or spray applied to the skin, or as an eye drop).
  • routes of administration including, for example, orally (for example, drenches as in aqueous or
  • the compound may also be formulated for inhalation.
  • a compound may be simply dissolved or suspended in sterile water. Details of appropriate routes of administration and compositions suitable for same can be found in, for example, U.S. Pat. Nos. 6,110,973, 5,763,493, 5,731,000, 5,541,231, 5,427,798, 5,358,970 and 4,172,896, as well as in patents cited therein.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, the particular mode of administration.
  • the amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.
  • Methods of preparing these formulations or compositions include the step of bringing into association an active compound, such as a compound provided herein, with the carrier and, optionally, one or more accessory ingredients.
  • an active compound such as a compound provided herein
  • the formulations are prepared by uniformly and intimately bringing into association a compound provided herein with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations provided herein suitable for oral administration may be in the form of capsules (including sprinkle capsules and gelatin capsules), cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), lyophile, powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound provided herein as an active ingredient.
  • Compositions or compounds may also be administered as a bolus, electuary or paste.
  • the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents,
  • pharmaceutically acceptable carriers such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose
  • compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres.
  • compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions that can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms useful for oral administration include pharmaceutically acceptable emulsions, lyophiles for reconstitution, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, cyclodextrins and derivatives thereof, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • Suspensions in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations of the pharmaceutical compositions for rectal, vaginal, or urethral administration may be presented as a suppository, which may be prepared by mixing one or more active compounds with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
  • suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
  • Formulations of the pharmaceutical compositions for administration to the mouth may be presented as a mouthwash, or an oral spray, or an oral ointment.
  • compositions can be formulated for delivery via a catheter, stent, wire, or other intraluminal device. Delivery via such devices may be especially useful for delivery to the bladder, urethra, ureter, rectum, or intestine.
  • Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants that may be required.
  • the ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to an active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound provided herein to the body.
  • dosage forms can be made by dissolving or dispersing the active compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.
  • Ophthalmic formulations eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this disclosure.
  • Exemplary ophthalmic formulations are described in U.S. Publication Nos. 2005/0080056, 2005/0059744, 2005/0031697 and 2005/004074 and U.S. Pat. No. 6,583,124, the contents of which are incorporated herein by reference.
  • liquid ophthalmic formulations have properties similar to that of lacrimal fluids, aqueous humor or vitreous humor or are compatible with such fluids.
  • a preferred route of administration is local administration (e.g., topical administration, such as eye drops, or administration via an implant).
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • compositions suitable for parenteral administration comprise one or more active compounds in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin.
  • the absorption of the drug in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microencapsulated matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissue.
  • active compounds can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • Methods of introduction may also be provided by rechargeable or biodegradable devices.
  • Various slow release polymeric devices have been developed and tested in vivo in recent years for the controlled delivery of drugs, including proteinacious biopharmaceuticals.
  • a variety of biocompatible polymers including hydrogels, including both biodegradable and non-degradable polymers, can be used to form an implant for the sustained release of a compound at a particular target site.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound or combination of compounds employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound(s) being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound(s) employed, the age, sex, weight, condition, general health and prior medical history of the subject being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the therapeutically effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the pharmaceutical composition or compound at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • therapeutically effective amount is meant the concentration of a compound that is sufficient to elicit the desired therapeutic effect. It is generally understood that the effective amount of the compound will vary according to the weight, sex, age, and medical history of the subject. Other factors which influence the effective amount may include, but are not limited to, the severity of the subject's condition, the disorder being treated, the stability of the compound, and, if desired, another type of therapeutic agent being administered with the compound provided herein.
  • a larger total dose can be delivered by multiple administrations of the agent.
  • Methods to determine efficacy and dosage are known to those skilled in the art (Isselbacher et al. (1996) Harrison's Principles of Internal Medicine 13 ed., 1814-1882, herein incorporated by reference).
  • a suitable daily dose of an active compound used in the compositions and methods provided herein will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • the effective daily dose of the active compound may be administered as one, two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • the active compound may be administered two or three times daily. In preferred embodiments, the active compound will be administered once daily.
  • compounds provided herein may be used alone or conjointly administered with another type of therapeutic agent.
  • the phrase “conjoint administration” refers to any form of administration of two or more different therapeutic compounds such that the second compound is administered while the previously administered therapeutic compound is still effective in the body (e.g., the two compounds are simultaneously effective in the subject, which may include synergistic effects of the two compounds).
  • the different therapeutic compounds can be administered either in the same formulation or in a separate formulation, either concomitantly or sequentially.
  • the different therapeutic compounds can be administered within one hour, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours, or a week of one another.
  • a subject who receives such treatment can benefit from a combined effect of different therapeutic compounds.
  • conjoint administration of compounds provided herein with one or more additional therapeutic agent(s) provides improved efficacy relative to each individual administration of the compound provided herein or the one or more additional therapeutic agent(s).
  • the conjoint administration provides an additive effect, wherein an additive effect refers to the sum of each of the effects of individual administration of the compound provided herein and the one or more additional therapeutic agent(s).
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water-soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal-chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. 5.5. THERAPEUTIC METHODS
  • provided herein are methods of treating cancer comprising administering a solid form or pharmaceutical composition provided herein. In one embodiment, provided herein are methods of using a solid form or pharmaceutical composition provided herein for treating, preventing or managing solid tumor. In one embodiment, provided herein is a method of treating solid tumor, comprising administering to a patient in need thereof a solid form or pharmaceutical composition provided herein.
  • the cancer is anaplastic large cell lymphoma (ALCL), atypical meningioma, breast cancer, cholangiocarcinoma, gastric cancer, glioblastoma, inflammatory myofibroblastic tumor (IMT), inflammatory hepatocellular adenoma (HCA), melanoma, pancreatic cancer, papillary thyroid carcinoma, salivary gland carcinoma, serous ovarian carcinoma, or spitzoid neoplasm.
  • ACL anaplastic large cell lymphoma
  • IMT inflammatory myofibroblastic tumor
  • HCA inflammatory hepatocellular adenoma
  • pancreatic cancer papillary thyroid carcinoma
  • salivary gland carcinoma salivary gland carcinoma
  • serous ovarian carcinoma or spitzoid neoplasm.
  • the solid tumor is advanced solid tumor. In one embodiment, the advanced solid tumor is relapsed after, refractory to, or resistant to the prior treatment by a tyrosine kinase inhibitor (TKI). In one embodiment, the solid tumor is non-small cell lung cancer (NSCLC). In one embodiment, the solid tumor is advanced NSCLC. In one embodiment, the solid tumor is locally advanced NSCLC. In one embodiment, the solid tumor is metastatic. In one embodiment, the solid tumor is CNS metastatic. In one embodiment, the solid tumor is metastatic NSCLC. In one embodiment, the solid tumor is CNS metastatic NSCLC. As used herein and unless otherwise specified, “advanced tumor” refers to a tumor that cannot be cured or grows beyond the initial site of origin, either locally advanced or metastatic.
  • the solid tumor (or cancer) is ROS1 positive. In one embodiment, the solid tumor is ROS1 positive NSCLC. In one embodiment, the solid tumor is advanced ROS1 positive solid tumor. In one embodiment, the solid tumor is locally advanced ROS1 positive solid tumor. In one embodiment, the solid tumor is advanced ROS1 positive NSCLC. In one embodiment, the solid tumor is locally advanced ROS1 positive NSCLC. In one embodiment, the solid tumor is metastatic ROS1 positive solid tumor. In one embodiment, the solid tumor is CNS metastatic ROS1 positive solid tumor. In one embodiment, the solid tumor is metastatic ROS1 positive NSCLC. In one embodiment, the solid tumor is CNS metastatic ROS1 positive NSCLC. In one embodiment, the solid tumor is CNS metastatic ROS1 positive NSCLC. In one embodiment, the solid tumor is CNS metastatic ROS1 positive NSCLC. In one embodiment, the solid tumor is CNS metastatic ROS1 positive NSCLC.
  • the solid tumor (or cancer) has a ROS1 mutation.
  • the ROS1 mutation is G2032R.
  • the ROS1 mutation comprise G2032R and one or more of S1986F, S1986Y, F2004C, F2004V, L2026M, D2033N, or G2101A.
  • the solid tumor (or cancer) has a ROS1 fusion.
  • the solid tumor (or cancer) is ALK positive.
  • an “ALK positive” (ALK+) cancer, disease, or disorder refers to a cancer, disease, or disorder characterized by inappropriately high expression of an ALK gene and/or the presence of a mutation in an ALK gene and/or the presence of a partially deleted ALK protein.
  • the solid tumor is ALK positive NSCLC.
  • the solid tumor is advanced ALK positive solid tumor.
  • the solid tumor is locally advanced ALK positive solid tumor.
  • the solid tumor is advanced ALK positive NSCLC.
  • the solid tumor is locally advanced ALK positive NSCLC.
  • the solid tumor is metastatic ALK positive solid tumor.
  • the solid tumor is CNS metastatic ALK positive solid tumor. In one embodiment, the solid tumor is metastatic ALK positive NSCLC. In one embodiment, the solid tumor is CNS metastatic ALK positive NSCLC. In one embodiment, the solid tumor (or cancer) has an ALK mutation.
  • the ALK mutation comprises one or more ALK rearrangements (in one embodiment, one rearrangement). In one embodiment, the ALK mutation comprises one or more ALK fusions (in one embodiment, one fusion). In some embodiments, cancers treated by methods of the present disclosure include ALK fusions. In one embodiment, the ALK fusion is with one of the fusion partners described in Ou et al., JTO Clinical and Research Reports, 1(1): 1-10, the entirety of which is incorporated herein by reference.
  • the ALK fusion is with one of the fusion partners selected from the group consisting of EML4, TFG, KIF5B, KLC1, STRN, HIP1, TPR, BIRC6, DCTN1, SQSTM1, SOCS5, SEC31A, CLTC, PRKAR1A, PPM1B, EIF2AK3, CRIM1, CEBPZ, PICALM, CLIP1, BCL11A, GCC2, LMO7, PHACTR1, CMTR1, VIT, DYSF, ITGAV, PLEKHA7, CUX1, VKORCIL1, FBXO36, SPTBN1, EML6, FBXO11, CLIP4, CAMKMT, NCOA1, MYT1L, SRBD1, SRD5A2, NYAP2, MPRIP, ADAM17, ALK, LPIN1, WDPCP, CEP55, ERC1, SLC16A7, TNIP2, ATAD2B, SLMAP, FBN1, SWAP70, TCF12, TRIM
  • the ALK fusion is with one of the fusion partners selected from the group consisting of EML4, TMP1, WDCP, GTF2IRD1, TPM3, TPM4, CLTC, LMNA, PRKAR1A, RANBP2, TFG, FN1, KLC1, VCL, STRN, HIP1, NPM1, DCTN1, SQSTM1, TPR, CRIM1, PTPN3, FBXO36, ATIC and KIF5B.
  • the ALK mutation is EML4-ALK, a fusion between the echinoderm microtubule-associated protein-like 4 (EML4) gene and the ALK tyrosine kinase domain.
  • the ALK mutation is NPM1-ALK. In one embodiment, the ALK mutation is STRN-ALK.
  • the solid tumor is leukocyte receptor tyrosine kinase (LTK) positive.
  • LTK positive LTK+
  • the solid tumor is LTK positive breast invasive ductal carcinoma, prostate adenocarcinoma, pancreatic adenocarcinoma, adenocarcinoma of unknown primary, or bladder urothelial carcinoma.
  • the cancer is LTK positive leukemia.
  • the solid tumor is LTK positive lung cancer. In one embodiment, the solid tumor is LTK positive NSCLC. In one embodiment, the solid tumor (or cancer) has an LTK mutation. In one embodiment, the LTK mutation is G269A, F218I, N257T, A13fs, or A214fs. In one embodiment, the solid tumor (or cancer) has an LTK fusion. In one embodiment, the LTK fusion is CLIP1-LTK. See Cooper A J, Sequist L V, Johnson T W, Lin J J. LTK fusions: A new target emerges in non-small cell lung cancer. Cancer Cell. 2022 Jan. 10; 40(1):23-25; and Izumi, H., Matsumoto, S., Liu, J. et al. The CLIP1-LTK fusion is an oncogenic driver in non-small-cell lung cancer. Nature 600, 319-323 (2021), each of which are incorporated herein by reference in their entirety.
  • the patient has not been treated with a prior therapy.
  • the patient is na ⁇ ve to (i.e. not receiving) any tyrosine kinase inhibitor (TKI) therapy.
  • TKI tyrosine kinase inhibitor
  • the patient has been treated with one or more prior therapies. In one embodiment, the patient has been treated with at least one prior TKI therapy. In one embodiment, the patient has been treated with at least two prior TKI therapies. In one embodiment, the patient has been treated with one prior TKI therapy. In one embodiment, the patient has been treated with two prior TKI therapies. In one embodiment, the patient's tumor does not have a known oncogenic driver alteration other than ROS1. In one embodiment, the TKI is ROS1 TKI (e.g. crizotinib or entrectinib). In one embodiment, the prior TKI therapy is one or more selected from the group consisting of crizotinib, entrectinib, repotrectinib, taletrectinib, and lorlatinib.
  • the prior TKI therapy is one or more selected from the group consisting of crizotinib, entrectinib, repotrectinib, taletrectinib, and lorlatinib.
  • the patient has not been treated with prior platinum-based chemotherapy. In one embodiment, the patient has been treated with up to one prior platinum-based chemotherapy. In one embodiment, the patient has been treated with at least one prior platinum-based chemotherapy. In one embodiment, the patient has been treated with at least two prior platinum-based chemotherapies. In one embodiment, the patient has been treated with one prior platinum-based chemotherapy. In one embodiment, the patient has been treated with two prior platinum-based chemotherapies.
  • platinum-based chemotherapy refers to chemotherapeutic agents that are coordination complexes of platinum.
  • Exemplified platinum-based chemotherapy includes but not limited to cisplatin, oxaliplatin, nedaplatin, or carboplatin.
  • the patient has not been treated with immunotherapy. In one embodiment, the patient has been treated with immunotherapy. In one embodiment, the patient has been treated with at least one prior immunotherapy. In one embodiment, the patient has been treated with at least two prior immunotherapies. In one embodiment, the patient has been treated with one prior immunotherapy. In one embodiment, the patient has been treated with two immunotherapies.
  • immunotherapy refers to the treatment of a disease by activating or suppressing the immune system. Immunotherapies designed to elicit or amplify an immune response are classified as activation immunotherapies, while immunotherapies that reduce or suppress are classified as suppression immunotherapies.
  • the immunotherapy can regulate the immune effector cells (e.g. lymphocytes, macrophages, dendritic cells, natural killer cells (NK Cell), cytotoxic T lymphocytes (CTL), etc.) to work together against cancer by targeting abnormal antigens expressed on the surface of tumor cells.
  • exemplified immunotherapy includes but not limited to checkpoint inhibitors (e.g.
  • Exemplified PD-1 inhibitors include but are not limited to pembrolizumab (Keytruda), nivolumab (Opdivo), and cemiplimab (Libtayo).
  • Exemplified PD-L1 inhibitors include but are not limited to atezolizumab (Tecentriq), avelumab (Bavencio), durvalumab (Imfinzi).
  • Exemplified CTLA-4 inhibitor includes but is not limited to ipilimumab (Yervoy).
  • the patient has not been treated with chemotherapy. In one embodiment, the patient has been treated with at least one prior line of chemotherapy. In one embodiment, the patient has been treated with at least two prior lines of chemotherapy.
  • the patient has been treated with at least three prior lines of anticancer therapy. In one embodiment, the patient has been treated with at least two prior lines of anticancer therapy selected from the group consisting of ROS1 TKI (e.g., investigational ROS1 TKI, crizotinib, lorlatinib, entrectinib, taletrectinib, repotrectinib) and chemotherapy. In one embodiment, the patient has been treated with at least one line of ROS1 TKI and one line of chemotherapy. In one embodiment, the patient has been treated with at least two lines of ROS1 TKI and one line of chemotherapy. In one embodiment, the patient has been treated with at least three lines of ROS1 TKI and one line of chemotherapy.
  • ROS1 TKI e.g., investigational ROS1 TKI, crizotinib, lorlatinib, entrectinib, taletrectinib, repotrectinib
  • chemotherapy e.g., investigational ROS1 TKI, crizot
  • the patient has been treated with at least two lines of chemotherapy. In one embodiment, the patient has been treated with at least one line of ROS1 TKI and two lines of chemotherapy. In one embodiment, the patient has been treated with at least two lines of ROS1 TKI and two lines of chemotherapy. In one embodiment, the patient has been treated with at least three lines of ROS1 TKI and two lines of chemotherapy.
  • the ROS1 TKI is crizotinib. In one embodiment, the ROS1 TKI is entrectinib. In one embodiment, the ROS1 TKI is lorlatinib. In one embodiment, the ROS1 TKI is repotrectinib. In one embodiment, the patient has been treated with lorlatinib and repotrectinib. In one embodiment, the ROS1 TKI is taletrectinib.
  • the solid tumor is metastatic ROS1-positive solid tumor, and the patient has been treated with at least one prior ROS1 TKI therapy.
  • the solid tumor is metastatic ROS1-positive NSCLC
  • the patient is na ⁇ ve to TKI therapy and has been treated with up to one prior platinum-based chemotherapy with or without immunotherapy.
  • the solid tumor is metastatic ROS1-positive NSCLC, and the patient has been treated with one prior ROS1 TKI therapy and has not been treated with prior platinum-based chemotherapy or immunotherapy.
  • the solid tumor is metastatic ROS1-positive NSCLC, and the patient has been treated with one prior ROS1 TKI therapy and one prior platinum-based chemotherapy with or without immunotherapy.
  • the solid tumor is metastatic ROS1-positive NSCLC, and the patient has been treated with at least two prior ROS1 TKI therapies and up to one prior platinum-based chemotherapy with or without immunotherapy.
  • the solid tumor is metastatic ROS1-positive solid tumor, and the patient has progressed on a prior therapy.
  • the prior therapy is a prior ROS1 TKI therapy.
  • the prior therapy is a prior chemotherapy (e.g., platinum-based chemotherapy).
  • the prior therapy is a prior immunotherapy.
  • the solid tumor is advanced ROS1-positive NSCLC
  • the patient is na ⁇ ve to TKI therapy and has been treated with up to one prior platinum-based chemotherapy with or without immunotherapy.
  • the solid tumor is advanced ROS1-positive NSCLC, and the patient has been treated with one prior ROS1 TKI therapy and has not been treated with prior platinum-based chemotherapy or immunotherapy.
  • the solid tumor is advanced ROS1-positive NSCLC, and the patient has been treated with one prior ROS1 TKI therapy and one prior platinum-based chemotherapy with or without immunotherapy.
  • the solid tumor is advanced ROS1-positive NSCLC, and the patient has been treated with at least two prior ROS1 TKI therapies and up to one prior platinum-based chemotherapy with or without immunotherapy.
  • the solid tumor is advanced ROS1-positive solid tumor, and the patient has progressed on a prior therapy.
  • the prior therapy is a prior ROS1 TKI therapy.
  • the ROS1 TKI is crizotinib. In one embodiment, the ROS1 TKI is entrectinib.
  • the prior therapy is a prior ROS1 TKI therapy.
  • the ROS1 TKI is crizotinib, entrectinib, taletrectinib, or repotrectinib.
  • the compound used herein (Compound 1 or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof) is administered once daily (QD). In one embodiment, the compound is administered twice daily (BID). In certain embodiments, the compound used herein is Compound 1.
  • the compound used herein (Compound 1 or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof) is administered at an amount of from about 5 mg to about 500 mg (by weight of the free base Compound 1) per day. In one embodiment, the compound is administered at an amount of from about 25 mg to about 250 mg per day. In one embodiment, the compound is administered at an amount of from about 25 mg to about 200 mg per day. In one embodiment, the compound is administered at an amount of from about 50 mg to about 200 mg per day. In one embodiment, the compound is administered at an amount of from about 5 mg to about 150 mg per day. In one embodiment, the compound is administered at an amount of from about 25 mg to about 150 mg per day.
  • the compound is administered at an amount of from about 25 mg to about 125 mg per day. In one embodiment, the compound is administered at an amount of from about 25 mg to about 100 mg per day. In one embodiment, the compound is administered at an amount of from about 50 mg to about 125 mg per day. In one embodiment, the compound is administered at an amount of from about 50 mg to about 100 mg per day.
  • the compound is administered at an amount of about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, or about 200 mg per day.
  • the amount is about 5 mg per day.
  • the amount is about 10 mg per day.
  • the amount is about 15 mg per day.
  • the amount is about 20 mg per day.
  • the amount is about 25 mg per day. In one embodiment, the amount is about 30 mg per day. In one embodiment, the amount is about 35 mg per day. In one embodiment, the amount is about 40 mg per day. In one embodiment, the amount is about 45 mg per day. In one embodiment, the amount is about 50 mg per day. In one embodiment, the amount is about 75 mg per day. In one embodiment, the amount is about 100 mg per day. In one embodiment, the amount is about 125 mg per day. In one embodiment, the amount is about 150 mg per day. As used herein, the weight amount refers to the weight amount of the free base Compound 1. In certain embodiments, the compound used herein is Compound 1.
  • the compound used herein (Compound 1 or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof) is administered at an amount of from about 5 mg to about 500 mg (by weight of the free base, Compound 1) once daily. In one embodiment, the compound is administered at an amount of from about 25 mg to about 250 mg once daily. In one embodiment, the compound is administered at an amount of from about 25 mg to about 200 mg once daily. In one embodiment, the compound is administered at an amount of from about 50 mg to about 200 mg once daily. In one embodiment, the compound is administered at an amount of from about 5 mg to about 150 mg once daily. In one embodiment, the compound is administered at an amount of from about 25 mg to about 150 mg once daily.
  • the compound is administered at an amount of from about 25 mg to about 125 mg once daily. In one embodiment, the compound is administered at an amount of from about 25 mg to about 100 mg once daily. In one embodiment, the compound is administered at an amount of from about 50 mg to about 125 mg once daily. In one embodiment, the compound is administered at an amount of from about 50 mg to about 100 mg once daily.
  • the compound is administered at an amount of about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, or about 200 mg once daily.
  • the amount is about 5 mg once daily.
  • the amount is about 10 mg once daily.
  • the amount is about 15 mg once daily.
  • the amount is about 20 mg once daily.
  • the amount is about 25 mg once daily. In one embodiment, the amount is about 30 mg once daily. In one embodiment, the amount is about 35 mg once daily. In one embodiment, the amount is about 40 mg once daily. In one embodiment, the amount is about 45 mg once daily. In one embodiment, the amount is about 50 mg once daily. In one embodiment, the amount is about 75 mg once daily. In one embodiment, the amount is about 100 mg once daily. In one embodiment, the amount is about 125 mg once daily. In one embodiment, the amount is about 150 mg once daily. As used herein, the weight amount refers to the weight amount of the free base Compound 1. In certain embodiments, the compound used herein is Compound 1.
  • the compound used herein (Compound 1 or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof) is administered at an amount of from about 5 mg to about 500 mg (by weight of the free base, Compound 1) twice daily. In one embodiment, the compound is administered at an amount of from about 5 mg to about 250 mg twice daily. In one embodiment, the compound is administered at an amount of from about 25 mg to about 250 mg twice daily. In one embodiment, the compound is administered at an amount of from about 25 mg to about 200 mg twice daily. In one embodiment, the compound is administered at an amount of from about 5 mg to about 100 mg twice daily. In one embodiment, the compound is administered at an amount of from about 10 mg to about 50 mg twice daily.
  • the compound is administered at an amount of from about 10 mg to about 100 mg twice daily. In one embodiment, the compound is administered at an amount of from about 25 mg to about 100 mg twice daily. In one embodiment, the compound is administered at an amount of from about 50 mg to about 100 mg twice daily. In one embodiment, the compound is administered at an amount of from about 5 mg to about 150 mg twice daily. In one embodiment, the compound is administered at an amount of from about 25 mg to about 150 mg twice daily.
  • the compound is administered at an amount of about 5, about 10, about 15, about 20, about 25, about 30, about 35, about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, or about 200 mg twice daily.
  • the amount is about 5 mg twice daily.
  • the amount is about 10 mg twice daily.
  • the amount is about 15 mg twice daily.
  • the amount is about 20 mg twice daily.
  • the amount is about 25 mg twice daily. In one embodiment, the amount is about 30 mg twice daily. In one embodiment, the amount is about 35 mg twice daily. In one embodiment, the amount is about 40 mg twice daily. In one embodiment, the amount is about 45 mg twice daily. In one embodiment, the amount is about 50 mg twice daily. In one embodiment, the amount is about 75 mg twice daily. In one embodiment, the amount is about 100 mg twice daily. In one embodiment, the amount is about 125 mg twice daily. In one embodiment, the amount is about 150 mg twice daily. As used herein, the weight amount refers to the weight amount of the free base Compound 1. In certain embodiments, the compound used herein is Compound 1.
  • the compound is administered orally.
  • the compound is administered in the form of one or more tablets.
  • the tablet has a unit dose strength of about 5 mg by weight of the free base Compound 1.
  • the table has a unit dose strength of about 25 mg by weight of the free base Compound 1.
  • the compound is administered to a subject with an empty stomach. In one embodiment, the compound is administered to a subject at fasted status. In one embodiment, the compound is administered to a subject without food. In one embodiment, the compound is administered to a subject at least 1 hour before and no sooner than 2 hours after ingestion of food and/or beverages other than water. In one embodiment, the compound is administered to a subject with a full stomach. In one embodiment, the compound is administered to a subject at fed status. In one embodiment, the compound is administered to a subject with food. In one embodiment, the compound is administered with the ingestion of food and/or beverages.
  • the patient experiences improvement in one or more symptoms selected from the group consisting of cognitive impairment, mood disorders, sleep disturbances, dizziness, ataxia, and weight gain, after the administration of the compound. In one embodiment, the patient does not experience one or more symptoms selected from the group consisting of cognitive impairment, mood disorders, sleep disturbances, dizziness, ataxia, and weight gain, after the administration of the compound. In one embodiment, the patient experiences reduced levels of one or more of pROS1, ROS1, pAKT, and pERK, after the administration of the compound. In one embodiment, the patient experiences reduced expression level of one or more MAP kinase pathway genes in tumor, after the administration of the compound.
  • the patient experiences reduced expression level of one or more MAP kinase pathway genes in solid tumor, after the administration of the compound.
  • the one or more MAP kinase pathway genes are selected from the group consisting of DUSP6, FOS, IL1R1, and SPRY4.
  • provided herein are methods of treating cancer comprising administering a therapeutically effective amount of a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • RTKs receptor tyrosine kinases
  • a family of membrane-bound proteins that transmit signals from outside the cell to promote cell survival, growth, and proliferation.
  • RTKs receptor tyrosine kinases
  • Aberrant RTK activation can lead to excessive cell growth and hence cancer.
  • RTKs contain an N-terminal domain that binds extracellular ligands, a transmembrane domain, and a C-terminal kinase domain that catalyzes intracellular signal transduction.
  • the compound of Formula (I) is an inhibitor of human ROS1.
  • ROS1 is an RTK encoded by the ROS1 gene.
  • the ligands and biological functions of human ROS1 are unknown, but its homologs in some other species have been shown to bind extracellular ligands and stimulate cell differentiation.
  • mouse ROS1 is essential for male gamete maturation and reproduction.
  • ROS1 chromosomal rearrangements are a well-documented cause of cancer, representing 1-2% of non-small cell lung cancer (NSCLC) and a subset of many other cancers. These rearrangements result in the fusion of the C-terminus of ROS1 with the N-terminus of various partner proteins, the most common of which is CD74.
  • NSCLC non-small cell lung cancer
  • ROS1 fusions have constitutive kinase activity that drives tumor growth through MAPK, PI3K, and JAK/STAT signaling pathways.
  • Small-molecule tyrosine kinase inhibitors (TKIs) have been used to target ROS1 fusions in cancer, including crizotinib and entrectinib.
  • Crizotinib was the first FDA-approved TKI for the treatment of ROS1-positive NSCLC. Despite an initial response, most patients acquire resistance to crizotinib and relapse. The predominant mechanism of resistance is the G2032R mutation in the solvent front, which dramatically reduces crizotinib affinity. No inhibitors with activity against ROS1-G2032R fusions have been FDA-approved, indicating a need in the art.
  • a compound provided herein selectively inhibits ROS1.
  • the compound selectively inhibits ROS1 over ALK.
  • the ratio of selectivity can be greater than a factor of about 1.5, greater than a factor of about 2, greater than a factor of about 3, greater than a factor of about 4, greater than a factor of about 5, greater than a factor of about 10, greater than a factor of about 20, greater than a factor of about 30, greater than a factor of about 50, or greater than a factor of about 100, where selectivity can be measured by ratio of IC 50 values, among other means.
  • the selectivity of ROS1 over ALK is measured by the ratio of the IC 50 value against ALK to the IC 50 value against ROS1.
  • the compound selectively inhibits ROS1 over TRK (e.g., TRKA, TRKB, and/or TRBC).
  • TRK e.g., TRKA, TRKB, and/or TRBC.
  • the ratio of selectivity can be greater than a factor of about 5, greater than a factor of about 10, greater than a factor of about 50, greater than a factor of about 100, greater than a factor of about 200, greater than a factor of about 400, greater than a factor of about 600, greater than a factor of about 800, greater than a factor of about 1000, greater than a factor of about 1500, greater than a factor of about 2000, greater than a factor of about 5000, greater than a factor of about 10,000, or greater than a factor of about 20,000, where selectivity can be measured by ratio of IC 50 values, among other means.
  • the selectivity of ROS1 over TRK is measured by the ratio of the IC 50 value against TRK to the IC 50 value against ROS1.
  • provided herein is a method for selectively inhibiting ROS1 over ALK wherein the inhibition takes place in a cell.
  • a method for selectively inhibiting ROS1 over TRK e.g., TRKA, TRKB, and/or TRBC
  • the method comprises contacting ROS1 with an effective amount of a solid form or a pharmaceutical composition provided herein.
  • such contact occurs in a cell.
  • such contact occurs in a cell in a mammal such as a human.
  • such contact occurs in a cell in human patient having a cancer provided herein.
  • provided herein is a method for selectively inhibiting ROS1 over ALK wherein the inhibition takes place in a subject suffering from cancer, said method comprising administering an effective amount of a solid form or a pharmaceutical composition provided herein to said subject.
  • a method of treating a subject suffering from a cancer associated with ROS1 said method comprising selectively inhibiting ROS1 over ALK by administering an amount of a solid form or a pharmaceutical composition provided herein to said subject, wherein said amount is sufficient for selective inhibiting ROS1 over ALK.
  • a method for selectively inhibiting ROS1 over TRK comprising administering an effective amount of a solid form or a pharmaceutical composition provided herein to said subject.
  • provided herein is a method of treating a subject suffering from a cancer associated with ROS1, said method comprising selectively inhibiting ROS1 over TRK (e.g., TRKA, TRKB, and/or TRBC) by administering an amount of a solid form or a pharmaceutical composition provided herein to said subject, wherein said amount is sufficient for selective inhibiting ROS1 over TRK (e.g., TRKA, TRKB, and/or TRBC).
  • TRK e.g., TRKA, TRKB, and/or TRBC
  • inhibition of ROS1 includes inhibition of wild type ROS1, or a mutation thereof
  • inhibition of ALK includes inhibition of wild type ALK, or a mutation thereof
  • inhibition of TRK includes inhibition of wild type TRK, or a mutation thereof.
  • Cancers treated by methods provided herein include, but are not limited to, lung cancer, e.g., non-small cell lung cancer, inflammatory myofibroblastic tumor, ovarian cancer, e.g., serous ovarian carcinoma, melanoma, e.g., spitzoid melanoma, glioblastoma, bile duct cancer, e.g., cholangiocarcinoma, gastric cancer, colorectal cancer, angiosarcoma, anaplastic large cell lymphoma, diffuse large B-cell lymphoma, large B-cell lymphoma, esophageal cancer, e.g., esophageal squamous cell carcinoma, kidney cancer, e.g., renal medullary carcinoma or renal cell carcinoma, breast cancer, e.g., triple negative breast cancer, thyroid cancer, e.g., papillary thyroid cancer, neuroblastoma, epithelioid hemangioend
  • Cancers treated by methods provided herein include cancers originating from one or more oncogenic proteins selected from ROS1, ALK, TRKA, TRKB, and TRKC. In certain embodiments, cancers treated by methods provided herein include cancers that are drug resistant to treatments directed at one or more oncogenic proteins selected from ROS1, ALK, TRKA, TRKB, and TRKC.
  • the cancer in a method provided herein is ROS1 positive (ROS1+).
  • ROS1 positive (ROS1+) cancer, disease, or disorder refers to a cancer, disease, or disorder characterized by inappropriately high expression of a ROS1 gene and/or the presence of a mutation in a ROS1 gene.
  • the mutation alters the biological activity of a ROS1 nucleic acid molecule or polypeptide.
  • a “mutation” or “mutant” of ROS1 comprises one or more deletions, substitutions, insertions, inversions, duplications, translocations, or amplifications in the amino acid or nucleotide sequences of ROS1, or fragments thereof.
  • a ROS1 “rearrangement” refers to genetic translocations involving the ROS1 gene that may result in ROS1 fusion genes and/or ROS1 fusion proteins.
  • the ROS1 fusion can also include one or more deletions, substitutions, insertions, inversions, duplications, translocations, or amplifications or a fragment thereof, as long as the mutant retains kinase phosphorylation activity.
  • the ROS1 mutation comprises one or more ROS1 point mutations.
  • cancers treated by methods provided herein include one or more mutations in ROS1 kinase.
  • the one or more ROS1 point mutations are selected from point mutations at E1935, L1947, L1951, G1971, E1974, L1982, S1986, F2004, E2020, L2026, G2032, D2033, C2060, F2075, L2086, V2089, V2098, G2101, D2113, 1981Tins, M2001T, and L2155.
  • the one or more ROS1 point mutations are selected from G2032R, G2032K, D2033N, S1986F, S1986Y, L2026M, L1951R, E1935G, L1947R, G1971E, E1974K, L1982F, F2004C, F2004V, E2020K, C2060G, F2075V, V2089M, V2098I, G2101A, D2113N, D2113G, L2155S, and L2086F.
  • the ROS1 mutation is G2032R.
  • the ROS1 mutation is S1986F.
  • the ROS1 mutation is S1986Y.
  • the ROS1 mutation is L2026M. In one embodiment, the ROS1 mutation is D2033N. In one embodiment, the ROS1 mutation is L2086F. In one embodiment, the ROS1 mutation is F2004C. In one embodiment, the ROS1 mutation is F2004V. In one embodiment, the ROS1 mutation is G2101A. In one embodiment, the ROS1 mutation is L1982F. In one embodiment, the ROS1 mutation is co-mutation of G2032R and one or more of S1986F, S1986Y, F2004C, F2004V, L2026M, or D2033N.
  • the ROS1 mutation comprises one or more ROS1 rearrangements (in one embodiment, one rearrangement). In one embodiment, the ROS1 mutation comprises one or more ROS1 fusions (in one embodiment, one fusion). In some embodiments, cancers treated by methods provided herein include ROS1 fusions.
  • the ROS1 fusion is with one of the fusion partners selected from SLC34A2, CD74, TPM3, SDC4, EZR, LRIG3, KDELR2, CEP72, CLTL, CTNND2, GOPC (e.g., GOPC-S, GOPC-L), GPRC6A, LIMA1, LRIG3, MSN, MYO5C, OPRM1, SLC6A17 SLMAP, SRSF6, TFG, TMEM106B, TPD52L1, ZCCHC8, CCDC6, CAPRIN1, CEP85L, CHCHD3, CLIP1, EEF1G, KIF21A, KLC1, SART3, ST13, TRIM24, ERC1, FIPIL1, HLAA, KIAA1598, MYO5A, PPFIBP1, PWWP2A, FN1, YWHAE, CCDC30, NCOR2, NFKB2, APOB, PLG, RBP4, and GOLGB1.
  • GOPC e.g., GOPC-S
  • the ROS1 fusion is CD74-ROS1 fusion. In one embodiment, the ROS1 fusion is SDC4-ROS1 fusion. In one embodiment, the ROS1 fusion is EZR-ROS1 fusion. In one embodiment, the ROS1 fusion is SLC34A2-ROS1 fusion. In one embodiment, the ROS1 fusion is GOPC-ROS1 fusion (e.g., GOPC-ROS1-S, GOPC-ROS1-L). In one embodiment, the ROS1 fusion is CEP85L-ROS1 fusion.
  • GOPC-ROS1 fusion e.g., GOPC-ROS1-S, GOPC-ROS1-L
  • the ROS1 fusion is CEP85L-ROS1 fusion.
  • the ROS1 mutation comprises one ROS1 rearrangement and one or more ROS1 point mutations.
  • the ROS1 mutation comprises one or more ROS1 rearrangements from CD74-ROS1, EZR-ROS1, SLC34A2-ROS1, GOPC-ROS1 (e.g., GOPC-ROS1-S, GOPC-ROS1-L), and CEP85L-ROS1, and one or more ROS1 point mutations selected from F2004C, F2004V, and G2032R.
  • the ROS1 mutation comprises one or more ROS1 rearrangements from CD74-ROS1, EZR-ROS1, and SLC34A2-ROS1, and ROS1 point mutation of G2101A.
  • the ROS1 mutation is CD74-ROS1 F2004C. In one embodiment, the ROS1 mutation is CD74-ROS1 F2004V. In one embodiment, the ROS1 mutation is CD74-ROS1 G2101A. In one embodiment, the ROS1 mutation is CD74-ROS1 G2032R. In one embodiment, the ROS1 mutation is CD74-ROS1 S1986F. In one embodiment, the ROS1 mutation is CD74-ROS1 L2026M. In one embodiment, the ROS1 mutation is CD74-ROS1 D2033N. In one embodiment, the ROS1 mutation is EZR-ROS1 F2004C. In one embodiment, the ROS1 mutation is EZR-ROS1 F2004V.
  • the ROS1 mutation is EZR-ROS1 G2101A. In one embodiment, the ROS1 mutation is EZR-ROS1 G2032R. In one embodiment, the ROS1 mutation is SLC34A2-ROS1 F2004C. In one embodiment, the ROS1 mutation is SLC34A2-ROS1 F2004V. In one embodiment, the ROS1 mutation is SLC34A2-ROS1 G2101A. In one embodiment, the ROS1 mutation is SLC34A2-ROS1 G2032R. In one embodiment, the ROS1 mutation is GOPC-ROS1 F2004C (e.g., GOPC-ROS1-S F2004C, GOPC-ROS1-L F2004C).
  • GOPC-ROS1 F2004C e.g., GOPC-ROS1-S F2004C, GOPC-ROS1-L F2004C.
  • the ROS1 mutation is GOPC-ROS1 F2004V (e.g., GOPC-ROS1-S F2004V, GOPC-ROS1-L F2004V).
  • the ROS1 mutation is GOPC-ROS1 G2032R (e.g., GOPC-ROS1-S G2032R, GOPC-ROS1-L G2032R).
  • the ROS1 mutation is CEP85L-ROS1 F2004C.
  • the ROS1 mutation is CEP85L-ROS1 F2004V.
  • the ROS1 mutation is CEP85L-ROS1 G2032R.
  • the ROS1 mutation is GOPC-ROS1 L1982F (e.g., GOPC-ROS1-S L1982F, GOPC-ROS1-L L1982F). In one embodiment, the ROS1 mutation is CD74-ROS1 L1982F.
  • the ROS1 cancer is determined by an FDA-approved test or other tests known in the art.
  • the tests that can be used include, e.g., OncomineTM Dx Target Test by Thermo Fisher Scientific. (a qualitative in vitro diagnostic test that uses targeted high-throughput, parallel-sequencing technology to detect sequence variations in 23 genes in DNA and RNA isolated from formalin-fixed, paraffin-embedded tumor (FFPE) tissue samples from patients with non-small cell lung cancer (NSCLC) using the Ion PGM Dx System); Vysis ROS1 Break Apart FISH Probe Kit (a qualitative test to detect rearrangements involving ROS1 gene rearrangements at 6q22 via fluorescence in situ hybridization (FISH) in formalin-fixed, paraffin-embedded (FFPE) non-small cell lung cancer (NSCLC) tissue specimens) or Reverse transcription-Polymerase Chain Reaction (RT-PCR) or Next Generation Sequencing (NGS) via a local diagnostic test.
  • FISH fluorescence in
  • Also provided are methods of treating a subject having a cancer e.g., a ROS1 positive cancer
  • a cancer e.g., a ROS1 positive cancer
  • methods of treating a subject having a cancer that include: determining whether a cancer cell in a sample obtained from a subject having a cancer and previously administered a first ROS1 inhibitor, has one or more ROS1 inhibitor resistance mutations; and administering a solid form or a pharmaceutical composition provided herein as a monotherapy or in conjunction with another anticancer agent to the subject if the subject has a cancer cell that has one or more ROS1 inhibitor resistance mutations.
  • the one or more ROS1 inhibitor resistance mutations confer increased resistance to a cancer cell or tumor to treatment with the first ROS1 inhibitor.
  • the one or more ROS1 inhibitor resistance mutations include one or more ROS1 inhibitor resistance mutations.
  • the one or more ROS1 inhibitor resistance mutations can include a substitution at one or more of amino acid positions 2032, 2033, 1986, 2026, 1951, 1935, 1947, 1971, 1974, 1982, 2004, 2020, 2060, 2075, 2089, 2098, 2101, 2113, 2155, 2032, and 2086, e.g., G2032R, D2033N, S1986F, S1986Y, L2026M, L1951R, E1935G, L1947R, G1971E, E1974K, L1982F, F2004C, F2004V, E2020K, C2060G, F2075V, V2089M, V2098I, G2101A, D2113N, D2113G, L2155S, L2032K, and L2086F.
  • another anticancer agent is any anticancer agent known in the art.
  • another anticancer agent can be another ROS1 inhibitor (e.g., a second ROS1 inhibitor).
  • a compound provided herein is a CNS-penetrating compound.
  • the compound after the administration of an effective amount of a solid form or a pharmaceutical composition provided herein (e.g., orally or intravenously), the compound is able to penetrate CNS (e.g., blood-brain barrier) and achieve a concentration in CNS (e.g., brain) that is still sufficient to inhibit (e.g., selectively inhibit) ROS1.
  • CNS e.g., blood-brain barrier
  • concentration in CNS e.g., brain
  • provided herein is a method for treating CNS metastases of a cancer, comprising administering to a subject in need thereof an effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • the CNS metastases is brain metastases.
  • the cancer is a ROS1+ cancer.
  • the compound is an inhibitor of human tropomyosin receptor kinase A, B, or C.
  • the IC 50 of the compound for inhibition of mutant or non-mutant ROS1 or ALK is no more than one-fifth of the IC 50 of the compound for inhibition of wild-type tropomyosin receptor kinase A, B, or C.
  • TRK inhibition particularly in the central nervous system (CNS), has been associated with adverse reactions, including dizziness/ataxia/gait disturbance, paraesthesia, weight gain and cognitive changes.
  • a method of minimizing adverse events in a subject in need of treatment for cancer comprising administering to the subject a therapeutically effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, and wherein the method minimizes adverse events associated with TRK inhibitors.
  • the cancer is a ROS1-associated cancer.
  • the adverse events are one or more of TRK-related CNS adverse events.
  • minimizing adverse events refers to a reduction in the incidence of adverse events in a subject or patient population compared to the paradigmatic incidence of adverse events in a subject or patient population treated with TRK inhibitors (e.g., entrectinib, repotrectinib, or lorlatinib).
  • the incidence of an adverse event refers to the frequency or percentage of a specific adverse event over a subject or patient population.
  • the incidence of an adverse event refers to the total number of adverse events experienced by an individual subject.
  • minimizing adverse events refers to minimizing TRK-related CNS adverse events.
  • minimizing TRK-related CNS adverse events means less than 40% of the patient population has a TRK-related CNS adverse event. In some embodiments, minimizing TRK-related CNS adverse events means less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10% or less than 5% of the patient population has a TRK-related CNS adverse event. In some embodiments, minimizing TRK-related CNS adverse events means less than 12% of the patient population have more than one TRK-related CNS adverse event.
  • minimizing TRK-related CNS adverse events means less than 11%, less than 10%, less than 9%, less than 8%, less than 7%, less than 6%, less than 5%, less than 4%, or less than 3% of the patient population have more than one TRK-related CNS adverse event.
  • TRK-related CNS adverse events refers to one or more of the following: dizziness, ataxia, gait disturbance, paraesthesia, weight gain, hyperphagia, paresthesias, abnormal movement, cognitive changes, speech effects (e.g, dysarthria, slow speech, or speech disorder), mood disorder (e.g., irritability, anxiety, depression, affect lability, personality change, mood swings, affective disorder, aggression, agitation, mood altered, depressed mood, euphoric mood, or mania), and cognitive disorder (e.g., memory impairment, cognitive disorder, amnesia, confusion, disturbance in attention, delirium, mental impairment, attention deficit/hyperactivity disorder, dementia, sleep disturbance, or reading disorder).
  • speech effects e.g, dysarthria, slow speech, or speech disorder
  • mood disorder e.g., irritability, anxiety, depression, affect lability, personality change, mood swings, affective disorder, aggression, agitation, mood altered, depressed mood, euphoric mood,
  • a method for preventing or limiting TRK-related CNS side effect or adverse event in a cancer treatment comprising administering to a subject in need thereof an effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • the method prevents the occurrence of the TRK-related CNS adverse event.
  • the method limits the frequency of occurrence of the TRK-related CNS adverse event.
  • the method limits the severity of the TRK-related side effect.
  • provided herein is a method for treating CNS metastases of a cancer with reduced TRK-related side effect, comprising administering to a subject in need thereof an effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • the reduction/limiting/prevention in CNS side effect or adverse event is determined in a statistical sample, as compared to a standard of care treatment, e.g., an approved ROS1 and/or ALK inhibitor (e.g., crizotinib, entrectinib, lorlatinib, or repotrectinib) for ROS1+ and/or ALK+ cancer.
  • a standard of care treatment e.g., an approved ROS1 and/or ALK inhibitor (e.g., crizotinib, entrectinib, lorlatinib, or repotrectinib) for ROS1+ and/or ALK+ cancer.
  • the TRK-related side effect is a TRKB-related CNS side effect.
  • the TRK-related CNS side effect or adverse event is dizziness, ataxia, gait disturbance, paraesthesia, weight gain, cognitive impairment, a mood disorder, or sleep disturbance.
  • a method for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • the cancer is a ROS1-associated cancer.
  • the cancer is a ROS1+ cancer.
  • the cancer is identified to be ROS1
  • provided herein is a method for treating a ROS1+ cancer, comprising administering to a subject in need thereof a therapeutically effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • a method for treating cancer in a subject comprising: (i) identifying the cancer in the subject to be ROS1+, and (ii) administering to the subject a therapeutically effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • the cancer (or ROS1+ cancer) is a solid tumor. In one embodiment, the cancer (or ROS1+ cancer) is an advanced solid tumor. In one embodiment, the cancer (or ROS1+ cancer) is a locally advanced solid tumor. In one embodiment, the cancer (or ROS1+ cancer) is lung cancer, e.g., non-small cell lung cancer (NSCLC), glioblastoma, inflammatory myofibroblastic tumor (IMT), bile duct cancer, e.g., cholangiocarcinoma, ovarian cancer, e.g., serous ovarian carcinoma, gastric cancer, colorectal cancer, angiosarcoma, melanoma, e.g., spitzoid melanoma, epithelioid hemangioendothelioma, esophageal cancer, e.g., esophageal squamous cell carcinoma (ESCC), kidney cancer, e.g., N
  • the cancer is lung cancer. In one embodiment, the cancer is non-small cell lung cancer. In one embodiment, the cancer is ROS1+ non-small cell lung cancer. In one embodiment, the cancer is relapsed or refractory non-small cell lung cancer. In one embodiment, the cancer is relapsed or refractory ROS1+ non-small cell lung cancer. In one embodiment, the cancer is newly diagnosed non-small cell lung cancer. In one embodiment, the cancer is newly diagnosed ROS1+ non-small cell lung cancer. In one embodiment, the cancer is bronchus cancer. In one embodiment, the cancer is ROS1+ bronchus cancer. In one embodiment, the cancer is relapsed or refractory bronchus cancer. In one embodiment, the cancer is relapsed or refractory ROS1+ bronchus cancer. In one embodiment, the cancer is newly diagnosed bronchus cancer. In one embodiment, the cancer is newly diagnosed ROS1+ bronchus cancer. In one embodiment, the cancer is newly diagnosed ROS1+ bronchus cancer
  • the cancer is glioblastoma. In one embodiment, the cancer is relapsed or refractory glioblastoma. In one embodiment, the cancer is relapsed or refractory ROS1+ glioblastoma. In one embodiment, the cancer is newly diagnosed glioblastoma. In one embodiment, the cancer is newly diagnosed ROS1+ glioblastoma.
  • the cancer is IMT. In one embodiment, the cancer is ROS1+ IMT. In one embodiment, the cancer is relapsed or refractory IMT. In one embodiment, the cancer is relapsed or refractory ROS1+ IMT. In one embodiment, the cancer is newly diagnosed IMT. In one embodiment, the cancer is newly diagnosed ROS1+ IMT.
  • the cancer is bile duct cancer. In one embodiment, the cancer is cholangiocarcinoma. In one embodiment, the cancer is ROS1+ cholangiocarcinoma. In one embodiment, the cancer is relapsed or refractory cholangiocarcinoma. In one embodiment, the cancer is relapsed or refractory ROS1+ cholangiocarcinoma. In one embodiment, the cancer is newly diagnosed cholangiocarcinoma. In one embodiment, the cancer is newly diagnosed ROS1+ cholangiocarcinoma.
  • the cancer is ovarian cancer. In one embodiment, the cancer is ROS1+ ovarian cancer. In one embodiment, the cancer is relapsed or refractory ovarian cancer. In one embodiment, the cancer is relapsed or refractory ROS1+ ovarian cancer. In one embodiment, the cancer is newly diagnosed ovarian cancer. In one embodiment, the cancer is newly diagnosed ROS1+ ovarian cancer. In one embodiment, the ovarian cancer is serous ovarian carcinoma. In one embodiment, the ovarian cancer is high grade serous ovarian carcinoma.
  • the cancer is gastric cancer. In one embodiment, the cancer is ROS1+ gastric cancer. In one embodiment, the cancer is relapsed or refractory gastric cancer. In one embodiment, the cancer is relapsed or refractory ROS1+ gastric cancer. In one embodiment, the cancer is newly diagnosed gastric cancer. In one embodiment, the cancer is newly diagnosed ROS1+ gastric cancer.
  • the cancer is colorectal cancer. In one embodiment, the cancer is ROS1+ colorectal cancer. In one embodiment, the cancer is relapsed or refractory colorectal cancer. In one embodiment, the cancer is relapsed or refractory ROS1+ colorectal cancer. In one embodiment, the cancer is newly diagnosed colorectal cancer. In one embodiment, the cancer is newly diagnosed ROS1+ colorectal cancer.
  • the cancer is angiosarcoma. In one embodiment, the cancer is ROS1+ angiosarcoma. In one embodiment, the cancer is relapsed or refractory angiosarcoma. In one embodiment, the cancer is relapsed or refractory ROS1+ angiosarcoma. In one embodiment, the cancer is newly diagnosed angiosarcoma. In one embodiment, the cancer is newly diagnosed ROS1+ angiosarcoma.
  • the cancer is melanoma. In one embodiment, the cancer is spitzoid tumor. In one embodiment, the cancer is spitzoid melanoma. In one embodiment, the cancer is ROS1+ spitzoid melanoma. In one embodiment, the cancer is relapsed or refractory spitzoid melanoma. In one embodiment, the cancer is relapsed or refractory ROS1+ spitzoid melanoma. In one embodiment, the cancer is newly diagnosed spitzoid melanoma. In one embodiment, the cancer is newly diagnosed ROS1+ spitzoid melanoma.
  • the cancer is epithelioid hemangioendothelioma. In one embodiment, the cancer is ROS1+ epithelioid hemangioendothelioma. In one embodiment, the cancer is relapsed or refractory epithelioid hemangioendothelioma. In one embodiment, the cancer is relapsed or refractory ROS1+ epithelioid hemangioendothelioma. In one embodiment, the cancer is newly diagnosed epithelioid hemangioendothelioma. In one embodiment, the cancer is newly diagnosed ROS1+ epithelioid hemangioendothelioma.
  • the cancer is esophageal cancer. In one embodiment, the cancer is ESCC. In one embodiment, the cancer is ROS1+ ESCC. In one embodiment, the cancer is relapsed or refractory ESCC. In one embodiment, the cancer is relapsed or refractory ROS1+ ESCC. In one embodiment, the cancer is newly diagnosed ESCC. In one embodiment, the cancer is newly diagnosed ROS1+ ESCC.
  • the cancer is kidney cancer. In one embodiment, the cancer is renal medullary carcinoma. In one embodiment, the cancer is ROS1+ renal medullary carcinoma. In one embodiment, the cancer is relapsed or refractory renal medullary carcinoma. In one embodiment, the cancer is relapsed or refractory ROS1+ renal medullary carcinoma. In one embodiment, the cancer is newly diagnosed renal medullary carcinoma. In one embodiment, the cancer is newly diagnosed ROS1+ renal medullary carcinoma. In one embodiment, the cancer is renal cell carcinoma. In one embodiment, the cancer is ROS1+ renal cell carcinoma. In one embodiment, the cancer is relapsed or refractory renal cell carcinoma. In one embodiment, the cancer is relapsed or refractory ROS1+ renal cell carcinoma. In one embodiment, the cancer is newly diagnosed renal cell carcinoma. In one embodiment, the cancer is newly diagnosed ROS1+ renal cell carcinoma. In one embodiment, the cancer is newly diagnosed ROS1+ renal cell carcinoma. In one embodiment, the cancer is newly diagnosed renal cell carcinoma.
  • the cancer is breast cancer. In one embodiment, the cancer is ROS1+ breast cancer. In one embodiment, the cancer is relapsed or refractory breast cancer. In one embodiment, the cancer is relapsed or refractory ROS1+ breast cancer. In one embodiment, the cancer is newly diagnosed breast cancer. In one embodiment, the cancer is newly diagnosed ROS1+ breast cancer. In one embodiment, the breast cancer is triple negative breast cancer.
  • the cancer is colon cancer. In one embodiment, the cancer is ROS1+ colon cancer. In one embodiment, the cancer is relapsed or refractory colon cancer. In one embodiment, the cancer is relapsed or refractory ROS1+ colon cancer. In one embodiment, the cancer is newly diagnosed colon cancer. In one embodiment, the cancer is newly diagnosed ROS1+ colon cancer.
  • the cancer is thyroid cancer. In one embodiment, the cancer is papillary thyroid cancer. In one embodiment, the cancer is ROS1+ papillary thyroid cancer. In one embodiment, the cancer is relapsed or refractory papillary thyroid cancer. In one embodiment, the cancer is relapsed or refractory ROS1+ papillary thyroid cancer. In one embodiment, the cancer is newly diagnosed papillary thyroid cancer. In one embodiment, the cancer is newly diagnosed ROS1+ papillary thyroid cancer.
  • the cancer is ROS1+ glioma (e.g. Grade 1, Grade 2, Grade 3, or Grade 4). In one embodiment, the cancer is relapsed or refractory glioma. In one embodiment, the cancer is relapsed or refractory ROS1+ glioma. In one embodiment, the cancer is newly diagnosed ROS1+ glioma. In one embodiment, the cancer is ROS1+ glioblastoma. In one embodiment, the cancer is newly diagnosed ROS1+ glioblastoma. In one embodiment, the cancer is relapsed or refractory glioblastoma. In one embodiment, the cancer is relapsed or refractory ROS1+ glioblastoma.
  • ROS1+ glioma e.g. Grade 1, Grade 2, Grade 3, or Grade 4
  • the cancer is relapsed or refractory glioma. In one embodiment, the cancer is relapsed or refractory ROS1+ glio
  • the cancer is neuroblastoma. In one embodiment, the cancer is ROS1+ neuroblastoma. In one embodiment, the cancer is relapsed or refractory neuroblastoma. In one embodiment, the cancer is relapsed or refractory ROS1+ neuroblastoma. In one embodiment, the cancer is newly diagnosed neuroblastoma. In one embodiment, the cancer is newly diagnosed ROS1+ neuroblastoma.
  • the cancer is ROS1+ pancreatic cancer. In one embodiment, the cancer is relapsed or refractory pancreatic cancer. In one embodiment, the cancer is relapsed or refractory ROS1+ pancreatic cancer. one embodiment, the cancer is newly diagnosed neuroblastoma. In one embodiment, the cancer is newly diagnosed ROS1+ pancreatic cancer.
  • the cancer is ROS1+ inflammatory hepatocellular adenoma. In one embodiment, the cancer is relapsed or refractory inflammatory hepatocellular adenoma. In one embodiment, the cancer is relapsed or refractory ROS1+ inflammatory hepatocellular adenoma. In one embodiment, the cancer is newly diagnosed neuroblastoma. In one embodiment, the cancer is newly diagnosed ROS1+ inflammatory hepatocellular adenoma.
  • the cancer is a hematological cancer.
  • the cancer or ROS1+ cancer, or ALK+ cancer
  • the lymphoma is non-Hodgkin lymphoma.
  • the lymphoma is anaplastic large cell lymphoma (ALCL), diffuse large B-cell lymphoma (DLBCL), or large B-cell lymphoma.
  • ACL aplastic large cell lymphoma
  • DLBCL diffuse large B-cell lymphoma
  • large B-cell lymphoma large B-cell lymphoma.
  • the cancer is ALCL. In one embodiment, the cancer is ROS1+ ALCL. In one embodiment, the cancer is ALK+ ALCL. In one embodiment, the cancer is relapsed or refractory ALCL. In one embodiment, the cancer is relapsed or refractory ROS1+ ALCL. In one embodiment, the cancer is relapsed or refractory ALK+ ALCL. In one embodiment, the cancer is newly diagnosed ALCL. In one embodiment, the cancer is newly diagnosed ROS1+ ALCL. In one embodiment, the cancer is newly diagnosed ALK+ ALCL.
  • the cancer is DLBCL. In one embodiment, the cancer is ROS1+ DLBCL. In one embodiment, the cancer is ALK+ DLBCL. In one embodiment, the cancer is relapsed or refractory DLBCL. In one embodiment, the cancer is relapsed or refractory ROS1+ DLBCL. In one embodiment, the cancer is relapsed or refractory ALK+ DLBCL. In one embodiment, the cancer is newly diagnosed DLBCL. In one embodiment, the cancer is newly diagnosed ROS1+ DLBCL. In one embodiment, the cancer is newly diagnosed ALK+ DLBCL.
  • the cancer is large B-cell lymphoma. In one embodiment, the cancer is ROS1+ large B-cell lymphoma. In one embodiment, the cancer is ALK+ large B-cell lymphoma. In one embodiment, the cancer is relapsed or refractory large B-cell lymphoma. In one embodiment, the cancer is relapsed or refractory ROS1+ large B-cell lymphoma. In one embodiment, the cancer is relapsed or refractory ALK+ large B-cell lymphoma. In one embodiment, the cancer is newly diagnosed large B-cell lymphoma. In one embodiment, the cancer is newly diagnosed ROS1+ large B-cell lymphoma. In one embodiment, the cancer is newly diagnosed ALK+ large B-cell lymphoma. In one embodiment, the cancer (or ROS1+ cancer) is new diagnosed. In one embodiment, the cancer (or ROS1+ cancer) is previously untreated.
  • the cancer (or ROS1+ cancer) is relapsed or refractory. In one embodiment, the cancer is relapsed. In one embodiment, the cancer (or ROS1+ cancer) is refractory.
  • the subject is previously untreated. In one embodiment, the subject is treatment na ⁇ ve to tyrosine kinase inhibitor (TKI) therapy. In one embodiment, the subject has received one or more prior lines of therapy. In one embodiment, the subject has received two or more prior lines of therapy. In one embodiment, the subject has developed resistance to one or more of the prior lines of therapy. In one embodiment, the prior therapy comprises a tyrosine kinase inhibitor (TKI).
  • TKI tyrosine kinase inhibitor
  • the prior TKI therapy comprises a treatment with one or more of crizotinib, ceritinib, alectinib, brigatinib, lorlatinib, entrectinib, repotrectinib, cabozantinib, foretinib, taletrectinib, merestinib, masitinib, and ensartinib.
  • the prior therapy comprises one or more chemotherapies.
  • the one or more chemotherapies are in addition to the TKI therapy.
  • the cancer is advanced cancer, e.g. relapsed after, refractory to, or resistant to the prior treatment by a TKI.
  • the cancer is resistant to a tyrosine kinase inhibitor (TKI).
  • TKI tyrosine kinase inhibitor
  • the cancer is resistant lung cancer. In one embodiment, the cancer is resistant non-small cell lung cancer. In one embodiment, the cancer is non-small cell lung cancer resistant to a TKI. In one embodiment, the cancer is ROS1+ non-small cell lung cancer resistant to a TKI.
  • the cancer is lung cancer (e.g., NSCLC).
  • the cancer is advanced lung cancer, e.g. relapsed after, or refractory to, prior treatment by a TKI.
  • a compound provided herein is administered as first-line treatment. In one embodiment, a compound provided herein is administered as second-line treatment. In one embodiment, a compound provided herein is administered as third or fourth-line treatment.
  • the cancer (or ROS1+ cancer) is metastatic. In one embodiment, the cancer has CNS metastases. In one embodiment, the cancer has brain metastases. In one embodiment, the cancer is metastatic non-small cell lung cancer (NSCLC). In one embodiment, the cancer is metastatic ROS1+ NSCLC.
  • NSCLC metastatic non-small cell lung cancer
  • a method for treating a patient with metastatic ROS1+ non-small cell lung cancer comprising administering to the patient a therapeutically effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • NSCLC metastatic ROS1+ non-small cell lung cancer
  • the patient is an adult patient. In one embodiment, the patient is a pediatric patient.
  • provided herein is a method for treating an adult patient with metastatic ROS1+ NSCLC, comprising administering to the patient a therapeutically effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • a method for treating an adult patient with metastatic ROS1+ NSCLC comprising administering to the patient a therapeutically effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, wherein the patient has progressed on or is intolerant of at least 1 prior TKI therapy.
  • a method for treating an adult patient with metastatic NSCLC that is ROS1+ with solvent front mutation G2032R comprising administering to the patient a therapeutically effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof, wherein the patient has progressed on or is intolerant of at least 1 prior TKI therapy.
  • TKI tyrosine kinase inhibitor
  • a method for treating a ROS1-associated (or ROS1+) cancer in a subject in need thereof, wherein the cancer has developed resistance to a tyrosine kinase inhibitor (TKI), and wherein the cancer has been identified as having one or more ROS1 inhibitor resistance mutations comprising administering to the subject a therapeutically effective amount of a solid form or a pharmaceutical composition provided herein, e.g., Form 1 of a compound of Formula (I), or a stereoisomer, or a mixture of stereoisomers thereof, or a pharmaceutically acceptable salt thereof.
  • TKI tyrosine kinase inhibitor
  • the one or more ROS1 inhibitor resistance mutations comprise one or more amino acid substitutions at an amino acid position selected from 1986, 2004, 2026, 2032, and 2033. In one embodiment, the one or more ROS1 inhibitor resistance mutations comprise one or more amino acid substitutions selected from S1986F, S1986Y, F2004C, F2004V, L2026M, G2032R, D2033N, L2086F, and G2101A. In one embodiment, the one or more ROS1 inhibitor resistance mutations is G2032R. In one embodiment, the one or more ROS1 inhibitor resistance mutations comprise G2032R and one or more of S1986F, S1986Y, F2004C, F2004V, L2026M, D2033N, or G2101A. In one embodiment, the ROS1 inhibitor resistance mutation is L2086F.
  • the TKI is a ROS1 inhibitor. In one embodiment, the TKI is an ALK inhibitor. In one embodiment, the TKI is crizotinib, ceritinib, alectinib, brigatinib, lorlatinib, entrectinib, repotrectinib, cabozantinib, foretinib, merestinib, taletrectinib, masitinib, or ensartinib. In one embodiment, the TKI is crizotinib. In one embodiment, the TKI is entrectinib.
  • the subject has relapsed after first-line treatment of the cancer. In other embodiments, the subject has relapsed after second-line treatment of the cancer.
  • the methods for treating or preventing cancer can be demonstrated by one or more responses such as increased apoptosis, inhibition of tumor growth, reduction of tumor metastasis, inhibition of tumor metastasis, reduction of microvessel density, decreased neovascularization, inhibition of tumor migration, tumor regression, and increased survival of the subject.
  • responses such as increased apoptosis, inhibition of tumor growth, reduction of tumor metastasis, inhibition of tumor metastasis, reduction of microvessel density, decreased neovascularization, inhibition of tumor migration, tumor regression, and increased survival of the subject.
  • the method of treating or preventing cancer may comprise administering a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), conjointly with one or more other chemotherapeutic agent(s).
  • the compound provided herein can be administered concurrently with, prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, 12 weeks, or 16 weeks before), or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, 12 weeks, or 16 weeks after), one or more other agents (e.g., one or more other additional agents).
  • each therapeutic agent is administered at a dose and/or on a time schedule determined for that particular agent.
  • the other therapeutic agent can be administered with the compound provided herein in a single composition or separately in a different composition. Triple therapy is also contemplated herein.
  • Chemotherapeutic agents that may be conjointly administered with the solid form or pharmaceutical composition provided herein include: 1-amino-4-phenylamino-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate (acid blue 25), 1-amino-4-[4-hydroxyphenyl-amino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-aminophenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[1-naphthylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-fluoro-2-carboxyphenylamino]-9,10-dioxo-9,10-dihydr
  • chemotherapeutic agents that may be conjointly administered with compounds provided herein include: ABT-263, dexamethasone, 5-fluorouracil, PF-04691502, romidepsin, and vorinostat (SAHA).
  • chemotherapeutic agents that may be conjointly administered with compounds provided herein include: 1-amino-4-phenylamino-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate (acid blue 25), 1-amino-4-[4-hydroxyphenyl-amino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-aminophenylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[1-naphthylamino]-9,10-dioxo-9,10-dihydroanthracene-2-sulfonate, 1-amino-4-[4-fluoro-2-carboxyphenylamino]-9,10-dioxo-9,10-dihydro
  • the chemotherapy agents are biologics such as ADCs or MET antibodies.
  • the chemotherapy agent is a MET inhibitor, a MEK inhibitor, a RET inhibitor, another ALK inhibitor, or ROS inhibitor.
  • the compound as described herein is used with one or more platininum based chemotherapy and/or immunotherapy (e.g. checkpoint inhibitors).
  • a solid form or pharmaceutical composition provided herein may be conjointly administered with one or more combination therapies.
  • Examples of combination therapies with which compounds provided herein may be conjointly administered are included in Table 1.
  • the conjoint therapies provided herein comprise conjoint administration with other types of chemotherapeutic agents, such as immuno-oncology agents.
  • Cancer cells often have specific cell surface antigens that can be recognized by the immune system.
  • immuno-oncology agents such as monoclonal antibodies, can selectively bind to cancer cell antigens and effect cell death.
  • Other immuno-oncology agents can suppress tumor-mediated inhibition of the native immune response or otherwise activate the immune response and thus facilitate recognition of the tumor by the immune system.
  • Exemplary antibody immuno-oncology agents include, but are not limited to, abagovomab, adecatumumab, afutuzumab, alemtuzumab, anatumomab mafenatox, apolizumab, blinatumomab, BMS-936559, catumaxomab, durvalumab, epacadostat, epratuzumab, indoximod, inotuzumab ozogamicin, intelumumab, ipilimumab, isatuximab, lambrolizumab, MED14736, MPDL3280A, nivolumab, obinutuzumab, ocaratuzumab, ofatumumab, olatatumab, pembrolizumab, pidilizumab, rituximab, ticilimumab, samalizum
  • the antibody immuno-oncology agents are selected from anti-CD73 monoclonal antibody (mAb), anti-CD39 mAb, anti-PD-1 mAb, and anti-CTLA4 mAb.
  • the methods provided herein comprise conjoint administration of one or more immuno-oncology agents, such as the agents mentioned above.
  • the combination therapy comprises conjoint administration of a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), with SH2 inhibitors, such as CGP78850, CPG85793, C90, C126, G7-18NATE, G7-B1, and NSC642056.
  • SH2 inhibitors such as CGP78850, CPG85793, C90, C126, G7-18NATE, G7-B1, and NSC642056.
  • the combination therapy comprises conjoint administration of a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), with ERK1/2 inhibitors such as ASN007, GDC-0994, KO-947, LTT462, LY3214996, MK-8353, ulixertinib.
  • the combination therapy comprises conjoint administration of a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), with MEK inhibitors, such as trametinib, cobimetinib, binimetinib, selumetinib, PD-325901, CI-1040, and TAK-733.
  • the combination therapy comprises conjoint administration of a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), with a MET inhibitor selected from JNJ-38877605, PF-04217903, foretinib, AMG 458, tivantinib, cabozantinib, crizotinib, capmatinib hydrochloride, tepotinib hydrochloride, and savolitinib.
  • a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I)
  • a MET inhibitor selected from JNJ-38877605, PF-04217903, foretinib, AMG 458, tivantinib, cabozantinib, crizotinib, capmatinib hydrochloride, tepotinib hydrochloride, and savolitinib.
  • the combination therapy comprises conjoint administration of solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), with a SHP2 inhibitor selected from TNO-155, RMC-4630, JAB-3068, or RLY-1971.
  • the combination therapy comprises conjoint administration of a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), with a RAS inhibitor selected from aliskiren, captopril, losartan, irbesartan, olmesartan, candesartan, valsartan, fimasartan, azilsartan, telmisartan, eprosartan, benazepril, enalapril, lisinopril, perindopril, quinapril, ramipril, and trandolapril.
  • a RAS inhibitor selected from aliskiren, captopril, losartan, irbesartan, olmesartan, candesartan, valsartan, fimasartan, azilsartan, telmisartan, eprosartan, benazepril, enalapril, lisinopril, perindopril
  • the combination therapy comprises administration of a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), in combination with a TKI.
  • the TKI is a ROS1 inhibitor.
  • the TKI is an ALK inhibitor.
  • the TKI is crizotinib, ceritinib, alectinib, brigatinib, lorlatinib, entrectinib, repotrectinib, cabozantinib, foretinib, merestinib, taletrectinib, masitinib, or ensartinib.
  • the TKI is crizotinib.
  • the TKI is entrectinib.
  • the TKI is alectinib.
  • the TKI is brigatinib.
  • the combination therapy comprises conjoint administration of a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I), with anti-PD-1 therapy.
  • the combination therapy comprises conjoint administration of a compound provided herein, such as a compound of Formula (I), with oxaliplatin.
  • the combination therapy comprises conjoint administration of a compound provided herein, such as a compound of Formula (I), with doxorubicin.
  • a solid form or pharmaceutical composition provided herein such as Form 1 of a compound of Formula (I) may be conjointly administered with non-chemical methods of cancer treatment.
  • a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I) may be conjointly administered with radiation therapy.
  • a solid form or pharmaceutical composition provided herein, such as Form 1 of a compound of Formula (I) may be conjointly administered with surgery, with thermoablation, with focused ultrasound therapy, with cryotherapy, or with any combination of these.
  • a solid form or pharmaceutical composition provided herein such as Form 1 of a compound of Formula (I) may be conjointly administered with one or more other compounds provided herein.
  • such combinations may be conjointly administered with other therapeutic agents, such as other agents suitable for the treatment of cancer, immunological or neurological diseases, such as the agents identified above.
  • conjointly administering one or more additional chemotherapeutic agents with a compound provided herein provides a synergistic effect.
  • conjointly administering one or more additional chemotherapeutic agents provides an additive effect.
  • Aqueous NaOH (9 L, 50 w %) was then added drop-wise to adjust the pH to 6-8 below 35° C.
  • the mixture was extracted with EtOAc (5V ⁇ 3), dried over Na 2 SO 4 (1.0 kg) and concentrated at 40-50° C. to provide crude Compound 11.
  • the wet filter cake was triturated in water (15 L) for 1-2 h at 20-25° C. and filtered again.
  • the filter cake was washed with water (5 L) and dried at 60° C. to afford Compound 9 as a yellow solid (4.66 kg, 98%/210 nm, 0.2% KF, 67% yield) with 1 H NMR features matching an authentic sample.
  • reaction mixture was washed with aqueous NaHCO 3 solution (2.4 kg, 20 kg water) at 10 ⁇ 20° C.
  • aqueous phase was extracted with DCM (7.5 L).
  • the combined organic phase was washed with water (12 L).
  • the organics were then concentrated under reduced pressure at 40 ⁇ 45° C. until no solvent distilled out.
  • MeCN (7.5 L) was added to get a solution of Compound 6, to which n-heptane (7.5 L) was added with stirring over 0.5 h at 15 ⁇ 25° C.
  • the upper n-heptane layer was removed, and the acetonitrile layer again extracted with n-heptane (7.5 L) with stirring for 0.5 h at 15 ⁇ 25° C.
  • n-heptane (3 vol. ⁇ 2) twice and concentrated at 40-45° C. to get the crude product as oil.
  • n-heptane (2 vol. relative to the crude product) was added, then heated to 40° C. and stirred for 1-2 h at 35-40° C. to get a clear solution.
  • the solution was slowly cooled to 15-20° C. over 1 h and stirred for 1-2 h at 15-20° C. Then the mixture was slowly cooled to 0° C. over 2 h and stirred at 0-5° C. for 16 h.
  • the organic phase was washed with cooled water (3.75 L) and separated.
  • the organic phase was dried over anhydrous Na 2 SO 4 , filtered and concentrated at 25-30° C. in vacuum to around 2 vol., then switched to n-heptane (2.25 L) and concentrated at 25-30° C. in vacuum to around 2 vol. of Compound 3 in n-heptane.
  • n-heptane/EtOAc (3.0 L, 10 v/1 v) was added to the above mixture and the mixture was slurried for 1 h at 0 ⁇ 10° C. under nitrogen and filtered. The filter cake was washed with n-heptane (1.5 L), dried in vacuum at 25 ⁇ 30° C.
  • n-heptane 950 mL, 1 vol. relative to crude product
  • n-heptane 950 mL, 1 vol. relative to crude product
  • the resulting mixture was cooled to 20-25° C. over 30 min and stirred for 1 h at 30-40° C.
  • n-heptane 1.9 L, 2 vol. relative to crude product
  • the precipitates were stirred at 15 ⁇ 20° C. for 3 h and filtered.
  • the filter cake was washed with n-heptane (1.5 L) and dried in oven at 45-50° C.
  • the solid was dissolved in EtOAc (600 mL, 2 vol.) at 50-60° C., then was added n-heptane (1.8 L, 6 vol.) dropwise over 50 min at 50-60° C. A large of solids came out during addition.
  • the resulting slurry was cooled to 15-20° C. over 50 min and stirred for 30 min at 15-20° C.
  • the slurry was concentrated in vacuum at 45-50° C. to 2-3 vol. mixture.
  • n-heptane (1.2 L, 4 vol.) was added to the above mixture (2-3 vol.), concentrated in vacuum at 45-50° C. to 2-3 vol. mixture. The mixture was cooled to 10 ⁇ 15° C. over 2 h, stirred at 10 ⁇ 15° C. for 1 h and filtered. The filtered cake was rinsed with n-heptane (600 mL) and dried in vacuum at 50° C. for 20 h to afford Form 1 of Compound 1 as an off-white solid (280 g, 99.0%).
  • a process was conducted in a manner analogous to that described above in the presence of about 0.02 equiv. of palladium acetate, 0.04 equiv. of CatacXiumA, 3.0 equiv. of potassium pivalate, and 8 vol. of t-amyl alcohol.
  • the crystallization was carried out from ethyl acetate/heptane and a slurrying process in toluene/heptane.
  • Form 2 was observed first from MTBE in the solvent solubility screen, and from the temperature cycled experiments. In this set of experiments, however, Form 2 was observed from a different solvent, 1-butanol, which indicates that Form 2 could be an isostructural solvate.
  • Form 4 was obtained from acetonitrile with heptane anti-solvent. Form 1 was also identified in the antisolvent addition experiments.
  • Form 1 A full crystal structure of Form 1 was collected and solved. See FIG. 6 for a representative depiction of the structure determined from single-crystal X-ray diffraction studies of Form 1.
  • a summary of structural data for Compound 1 Form 1 is provided in the table below.
  • R1 ( ⁇
  • the asymmetric unit of Form 1 contains one fully ordered molecule of Compound 1.
  • Anisotropic atomic displacement ellipsoids for the non-hydrogen atoms are shown at the 5000 probability level. Hydrogen atoms are displayed with an arbitrarily small radius.
  • Single crystals of Form 7 were obtained by first slurrying 500 mg of amphorus Compound 1 in MTBE. The sample was temperature cycled in an incubator shaker between ambient and 40° C. over 4 hour cycles for ca. 20 hours, at which point XRPD confirmed that the material had converted to Form 2. Ca. 100 mg of Form 2 material was placed in a 5 ml scintillation vial in a Büchi glass oven. The sample was heated up to 250° C. at a heating rate of 20° C./min. The sample was held at 250° C. for 6 min and then cooled down to room temperature as quickly as possible using wet paper towels and acetone to aid with the cooling. An aliquot was analyzed by XRPD and confirmed formation of Form 7.
  • Form 7 A full crystal structure of Form 7 was collected and solved. See FIG. 23 for a representative depiction of the structure determined from single-crystal X-ray diffraction studies of Form 1.
  • a summary of structural data for Compound 1 Form 7 is provided in the table below.
  • Flack parameter 0.01(3) ( ⁇
  • the asymmetric unit of Form 7 contains one fully ordered molecule of Compound 1.
  • Anisotropic atomic displacement ellipsoids for the non-hydrogen atoms are shown at the 50% probability level. Hydrogen atoms are displayed with an arbitrarily small radius.
  • Form 1 of Compound 1 Method. About 10 mg of Form 1 of Compound 1 was placed in a 2 ml vial and stored for 7 days under the following conditions: A) 40° C./75% RH (open vial); B) 25° C./60% RH (open vial); C) 80° C. (sealed vial). Solids were analyzed by XRPD and submitted for HPLC.
  • Form 1 of Compound 1 Results. About HPLC analysis of the three samples of Compound 1 pattern 1 after storage at 40° C./75% RH (open vial), 25° C./60% RH (open vial), and 80° C. (sealed vial), for one week, showed high purity (>99.5%) in all the samples. HPLC analysis results of the 7-days stability studies performed on Form 1 of Compound 1 can be found in the table below. Form 1 is stable under all tested conditions.
  • Form 2 of Compound 1 Method. Portions of Form 1 of Compound 1 (10 mg) were placed in 1.5 ml vials. One vial was stored for 7 days under each of the following conditions: A) 40° C./75% RH (open vial); B) 25° C./60% RH (open vial); C) 80° C. (sealed vial). Solids were analyzed by XRPD and submitted for HPLC.
  • Form 2 of Compound 1 Results. Scaled up Form 2 was determined to be stable at 40° C./75% RH and 25° C./60% RH for 7 days as the XRPD pattern was observed to correspond to that of Form 2. However, for sample C stored at 80° C., the XRPD pattern was observed to correspond to that for Form 7.
  • Modulated DSC thermograms of Form 1 after 100 MPa compression test sample showed an endothermic event in the non-reversing heat flow trace with an onset temperature of ca. 265° C., and an endothermic event in the reversing heat flow trace with an onset temperature of ca. 268° C. No exothermic events were observed in any of the traces for this sample. There was no evidence of amorphous content in the material.
  • Modulated DSC thermograms of Form 1 after 250 MPa compression test sample showed an endothermic event in the non-reversing heat flow trace with an onset temperature of ca. 266° C., and an endothermic event in the reversing heat flow trace with an onset temperature of ca. 266° C. No exothermic events were observed in any of the traces for this sample. There was no evidence of amorphous content in the material.
  • Form A of a phosphate salt of Compound 1 identified from the primary salt screen was analyzed by XRPD ( FIG. 28 ).
  • the 1 H NMR and 31 P NMR spectra of Form A of a phosphate salt of Compound 1 confirmed salt formation (e.g. 31 F signal observed at about 0.9 ppm) and retention of the bonding connectivity of Compound 1.
  • NMR experiments were performed on a Bruker AVIIIHD spectrometer equipped with a PRODIGY cryoprobe operating at 500.23 MHz for protons. Experiments were performed in deuterated dimethyl sulfoxide and each sample was prepared to ca. 10 mM concentration.
  • Infrared spectroscopy was carried out on a Bruker ALPHA P spectrometer. Sufficient material was placed onto the centre of the plate of the spectrometer and the spectra were obtained using the following parameters: Resolution: 4 cm ⁇ 1 ; Background Scan Time: 16 scans; Sample Scan Time: 16 scans; Data Collection: 4000 to 400 cm-1; Result Spectrum: Transmittance; Software: OPUS version 6.
  • sample was placed into a mesh vapour sorption balance pan and loaded into a DVS Advantage dynamic vapour sorption balance by Surface Measurement Systems.
  • the sample was subjected to a ramping profile from 40-90% relative humidity (RH) at 10% increments, maintaining the sample at each step until a stable weight had been achieved (dm/dt 0.004%, minimum step length 30 minutes, maximum step length 500 minutes) at 25° C.
  • RH relative humidity
  • the sample was dried using the same procedure to 0% RH and then a second sorption cycle back to 40% RH. Two cycles were performed. The weight change during the sorption/desorption cycles were plotted, allowing for the hygroscopic nature of the sample to be determined. XRPD analysis was then carried out on any solid retained.
  • Compound 1 drug substance and each excipient are weighed and dispensed according to the batch formula.
  • Intragranular excipients mannitol, silicified microcrystalline cellulose, hydroxypropyl cellulose, and sodium starch glycolate
  • Intragranular excipients mannitol, silicified microcrystalline cellulose, hydroxypropyl cellulose, and sodium starch glycolate
  • Magnesium stearate is screened, combined with the pre-granulation blend, and blended.
  • the resulting final pre-granulation blend is then fed through a roller compactor and comil to generate Compound 1 granules.
  • the Compound 1 granules are screened with the extragranular excipients (mannitol and sodium starch glycolate) and blended.
  • Magnesium stearate is screened, combined with the extragranular blend, and blended to achieve the Compound 1 final blend.
  • the Compound 1 final blend is transferred to an automated tablet press set up to manufacture Compound 1 tablets in the appropriate shape, size, and weight. Throughout the compression run tablets are sampled and tested for weight, appearance, thickness, hardness, and friability. The resulting tablets are passed through a deduster and metal checker prior to film coating.
  • An aqueous suspension of Opadry® Pink II is prepared and sprayed onto the tablets in a pan-coater until the target weight gain is achieved. Mean tablet weights are monitored throughout the coating process to ensure proper weight control and achieve the coating endpoint.
  • a roller compacted batch of Compound 1 was manufactured with the formula and conditions in the following table.
  • Stability results demonstrated the chemical and physical stability of Compound 1 stored for 12 months at the proposed long-term condition of 25° C. ⁇ 2° C./60% ⁇ 5% RH and 6 months at the accelerated condition of 40° C. ⁇ 2° C./75% ⁇ 5% RH.
  • No meaningful changes were observed in description, assay, related substances, water content, polymorphic form, and chiral purity. All results complied with the acceptance criteria (e.g. drug substance purity: at least 97%, individual impurities: each less than or equal to 0.2% by area, total amount of impurities: less than or equal to 2.0%, enantiomeric purity: at least 99.5%, solid form: Form 1) at all time points.
  • Compound 1 and each excipient are weighed and dispensed according to the batch formula.
  • Mannitol, silicified microcrystalline cellulose, hydroxypropyl cellulose, and sodium starch glycolate are screened with the Compound 1 through a 20-mesh screen to delump and subsequently blended to form an initial blend.
  • Magnesium stearate is screened and combined with the initial blend and blended to achieve a final blend.
  • the final blend is transferred to an automated tablet press set up to manufacture tablets in the appropriate shape, size, and weight. Throughout the compression run tablets are sampled and tested for weight, appearance, thickness, hardness, and friability. The resulting tablets are passed through a deduster and metal checker prior to film coating.
  • An aqueous suspension of Opadry® Pink II is prepared and sprayed onto the tablets in a pan-coater until the target weight gain (40%) is achieved. Mean tablet weights are monitored throughout the coating process to ensure proper weight control and achieve the coating endpoint.
  • the formulation is designed as dose proportional. Common blend is compressed into tablets with different strengths (e.g. 25 mg, 50 mg, 75 mg and 100 mg). Representative composition and formulation of tablets of Compound 1 is listed in the following table.
  • Results demonstrate the chemical and physical stability of tablets of Compound 1 upon storage for up to 12 months under the long-term ICH condition of 25° C. 2° C./60% ⁇ 5% RH and under the ICH intermediate condition of 30° C. ⁇ 2° C./65% 5% RH as well as 6 months under the ICH accelerated condition of 40° C. ⁇ 2° C./75% ⁇ 5% RH. No meaningful changes were observed in description, assay and degradation products, chiral purity, water content, and dissolution.
  • Results demonstrate the chemical and physical stability of tablets of Compound 1 upon storage for 12 months under long-term ICH condition of 30° C. ⁇ 2° C./65% ⁇ 5% RH as well as 6 months under the ICH accelerated condition of 40° C. ⁇ 2° C./75% ⁇ 5% RH. No meaningful changes were observed in description, assay and degradation products, water content, and dissolution.
  • Results for Tables Prepared as Described in Example 13 Results demonstrate the chemical and physical stability of tablets of Compound 1 upon storage for up to 6 months under long-term ICH conditions of 30° C. ⁇ 2° C./65% ⁇ 5% RH as well as ICH accelerated condition of 40° C. ⁇ 2° C./75% ⁇ 5% RH. No meaningful changes were observed in description, assay and degradation products, chiral purity, water content, and dissolution.
  • Results demonstrate the chemical and physical stability of tablets of Compound 1 upon storage for 3 months under long-term ICH condition of 30° C. ⁇ 2° C./65% ⁇ 5% RH as well as ICH accelerated condition of 40° C. 2° C./75% ⁇ 5% RH. No meaningful changes were observed in description, assay and degradation products, water content, and dissolution.
  • a wet mill may be employed during crystallization of Compound 1 in order to control the particle size distribution of the formed material.
  • an IKA Works wet mill furnished with a 2P/4M rotor stator stack, is hooked up in line to the process tank. The wet mill is run at a speed such that the desired particle size (e.g., D50 ⁇ 50-100 microns, D10>10 microns, D90 ⁇ 150 microns) can be achieved after approximately 25 passes through the wet mill.
  • the amount of milling time can be made shorter, or longer, by changing the rpms at which the mill operates at. It is important to note that the material is friable in nature, and can be over-milled if the wrong setting is chosen.
  • Particle Size for milled samples is determined by filtering a sample from the reaction mixture, and suspending the sample in an appropriate dispersant.
  • the appropriate dispersant for Compound 1 is water with a small amount of additional surfactant, such as Triton X.
  • the material can then be measured for its particle size distribution using an instrument such as a Malvern Mastersizer 3000, or something analogous.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US17/957,725 2021-10-01 2022-09-30 Solid forms, pharmaceutical compositions and preparation of heteroaromatic macrocyclic ether compounds Pending US20230124705A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/957,725 US20230124705A1 (en) 2021-10-01 2022-09-30 Solid forms, pharmaceutical compositions and preparation of heteroaromatic macrocyclic ether compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163251514P 2021-10-01 2021-10-01
US17/957,725 US20230124705A1 (en) 2021-10-01 2022-09-30 Solid forms, pharmaceutical compositions and preparation of heteroaromatic macrocyclic ether compounds

Publications (1)

Publication Number Publication Date
US20230124705A1 true US20230124705A1 (en) 2023-04-20

Family

ID=84044639

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/957,725 Pending US20230124705A1 (en) 2021-10-01 2022-09-30 Solid forms, pharmaceutical compositions and preparation of heteroaromatic macrocyclic ether compounds

Country Status (6)

Country Link
US (1) US20230124705A1 (zh)
AU (1) AU2022357554A1 (zh)
CA (1) CA3231813A1 (zh)
IL (1) IL311444A (zh)
TW (1) TW202320768A (zh)
WO (1) WO2023056405A1 (zh)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024036097A1 (en) 2022-08-12 2024-02-15 Nuvalent, Inc. Heteroaromatic macrocyclic ether compounds and isotopologues thereof

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4172896A (en) 1978-06-05 1979-10-30 Dainippon Pharmaceutical Co., Ltd. Methane-sulfonamide derivatives, the preparation thereof and composition comprising the same
GB9217295D0 (en) 1992-08-14 1992-09-30 Wellcome Found Controlled released tablets
US5358970A (en) 1993-08-12 1994-10-25 Burroughs Wellcome Co. Pharmaceutical composition containing bupropion hydrochloride and a stabilizer
US5541231A (en) 1993-07-30 1996-07-30 Glaxo Wellcome Inc. Stabilized Pharmaceutical
GB9315856D0 (en) 1993-07-30 1993-09-15 Wellcome Found Stabilized pharmaceutical
CN1229110C (zh) 1997-07-29 2005-11-30 阿尔康实验室公司 含半乳甘露聚糖聚合物和硼酸盐的眼用组合物
WO1999038504A1 (en) 1998-01-29 1999-08-05 Sepracor Inc. Pharmaceutical uses of optically pure (-)-bupropion
US8889112B2 (en) 1999-09-16 2014-11-18 Ocularis Pharma, Llc Ophthalmic formulations including selective alpha 1 antagonists
US6933289B2 (en) 2003-07-01 2005-08-23 Allergan, Inc. Inhibition of irritating side effects associated with use of a topical ophthalmic medication
ATE444732T1 (de) 2003-08-07 2009-10-15 Allergan Inc Zusammensetzungen zur abgabe von therapeutika in die augen und verfahren zu ihrer herstellung und verwendung
US20050059744A1 (en) 2003-09-12 2005-03-17 Allergan, Inc. Methods and compositions for the treatment of pain and other alpha 2 adrenergic-mediated conditions
CN104169286B (zh) * 2012-03-06 2016-06-08 辉瑞大药厂 用于治疗增殖性疾病的大环衍生物
CN115916754A (zh) * 2020-05-05 2023-04-04 纽威伦特公司 杂芳族大环醚化学治疗剂

Also Published As

Publication number Publication date
CA3231813A1 (en) 2023-04-06
TW202320768A (zh) 2023-06-01
IL311444A (en) 2024-05-01
WO2023056405A1 (en) 2023-04-06
AU2022357554A1 (en) 2024-04-11

Similar Documents

Publication Publication Date Title
JP6946194B2 (ja) キナーゼを調節する化合物の固体形態
AU2017419352B2 (en) Pharmaceutical compositions
KR101829595B1 (ko) 3-(1-{3-[5-(1-메틸-피페리딘-4일메톡시)-피리미딘-2-일]-벤질}-6-옥소-1,6-디히드로-피리다진-3-일)-벤조니트릴 히드로클로라이드 염의 신규한 다형체 및 이의 제조 방법
AU2017335242C1 (en) Pyridine compound
TW201945357A (zh) 化合物
KR20190003750A (ko) 암을 치료하기 위한 1-테트라히드로피라닐카르보닐-2,3-디히드로-1h-인돌 화합물
TW201718513A (zh) 製備經取代之3-(2-苯胺-1-環己基-1h-苯并咪唑-5-基)丙酸衍生物之方法
WO2004037812A1 (ja) 複素環式化合物及びそれを有効成分とする抗腫瘍剤
JP2021517902A (ja) 置換ピロロトリアジン系化合物およびその医薬組成物並びにそれらの使用
US20140018372A1 (en) Crystalline form of a indolinone derivative and its use
US20230124705A1 (en) Solid forms, pharmaceutical compositions and preparation of heteroaromatic macrocyclic ether compounds
TW200914442A (en) Gamma secretase modulators
WO2007032371A1 (ja) 摂食調節剤としてのオキシインドール誘導体
AU2017326297B2 (en) 7-substituted 1-aryl-naphthyridine-3-carboxylic acid amides and use thereof
WO2020224626A9 (zh) 用作激酶抑制剂的化合物及其应用
US20210032218A1 (en) Succinate forms and compositions of bruton's tyrosine kinase inhibitors
TW200408392A (en) Crystalline 2,5-dione-3-(1-methyl-1H-indol-3-yl)-4-[1-(pyridin-2-ylmethyl )piperidin-4-yl]-1H-indol-3-yl]-1H-pyrrole mono -hydrochiloride
WO2022095910A1 (zh) 用作激酶抑制剂的化合物及其应用
CN109843300B (zh) 一种化合物的结晶多晶型形式
TW201932117A (zh) 經取代三唑衍生物之前驅藥及其用途
CA3231608A1 (en) Methods of treating solid tumor using heteroaromatic macrocyclic ether compounds
US20230322797A1 (en) Solid forms, pharmaceutical compositions and preparation of heteroaromatic macrocyclic ether compounds
WO2011088075A2 (en) Novel salts, polymorphs, and synthetic processes regarding imidazole derivative
WO2024036097A1 (en) Heteroaromatic macrocyclic ether compounds and isotopologues thereof
JP2015516440A (ja) 7−(tert−ブチル−d9)−3−(2,5−ジフルオロフェニル)−6−((1−メチル−1H−1,2,4−トリアゾール−5−イル)メトキシ)−[1,2,4]トリアゾロ[4,3−b]ピリダジンの2型多形

Legal Events

Date Code Title Description
AS Assignment

Owner name: CAMBREX CORPORATION, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PEARSON, DAVID JAMES;REEL/FRAME:061528/0937

Effective date: 20221011

Owner name: NUVALENT, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GERARD, BAUDOUIN;HORAN, JOSHUA COURTNEY;KROPP, JASON T.;AND OTHERS;SIGNING DATES FROM 20221006 TO 20221018;REEL/FRAME:061528/0924

Owner name: NUVALENT, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CAMBREX CORPORATION;REEL/FRAME:061528/0914

Effective date: 20221012

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: NUVALENT, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CHEN, SIBAO;COOPER, CHRISTOPHER G. F.;SIGNING DATES FROM 20221203 TO 20230105;REEL/FRAME:064020/0247

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS